CN110337448B - Immunotherapy using antibodies that bind to programmed death ligand 1 (PD-L1) - Google Patents

Immunotherapy using antibodies that bind to programmed death ligand 1 (PD-L1) Download PDF

Info

Publication number
CN110337448B
CN110337448B CN201780087354.6A CN201780087354A CN110337448B CN 110337448 B CN110337448 B CN 110337448B CN 201780087354 A CN201780087354 A CN 201780087354A CN 110337448 B CN110337448 B CN 110337448B
Authority
CN
China
Prior art keywords
seq
antibody
light chain
heavy chain
variable region
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Active
Application number
CN201780087354.6A
Other languages
Chinese (zh)
Other versions
CN110337448A (en
Inventor
弗兰克·J·卡尔佐内
严海
约翰·张
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Remd Biotherapeutics Inc
Original Assignee
Remd Biotherapeutics Inc
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Remd Biotherapeutics Inc filed Critical Remd Biotherapeutics Inc
Publication of CN110337448A publication Critical patent/CN110337448A/en
Application granted granted Critical
Publication of CN110337448B publication Critical patent/CN110337448B/en
Active legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/2803Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the immunoglobulin superfamily
    • C07K16/2827Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the immunoglobulin superfamily against B7 molecules, e.g. CD80, CD86
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/39Medicinal preparations containing antigens or antibodies characterised by the immunostimulating additives, e.g. chemical adjuvants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/395Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum
    • A61K39/39533Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum against materials from animals
    • A61K39/3955Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum against materials from animals against proteinaceous materials, e.g. enzymes, hormones, lymphokines
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/005Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies constructed by phage libraries
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K19/00Hybrid peptides, i.e. peptides covalently bound to nucleic acids, or non-covalently bound protein-protein complexes
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/63Introduction of foreign genetic material using vectors; Vectors; Use of hosts therefor; Regulation of expression
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N5/00Undifferentiated human, animal or plant cells, e.g. cell lines; Tissues; Cultivation or maintenance thereof; Culture media therefor
    • C12N5/10Cells modified by introduction of foreign genetic material
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/20Immunoglobulins specific features characterized by taxonomic origin
    • C07K2317/24Immunoglobulins specific features characterized by taxonomic origin containing regions, domains or residues from different species, e.g. chimeric, humanized or veneered
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/50Immunoglobulins specific features characterized by immunoglobulin fragments
    • C07K2317/55Fab or Fab'
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/50Immunoglobulins specific features characterized by immunoglobulin fragments
    • C07K2317/56Immunoglobulins specific features characterized by immunoglobulin fragments variable (Fv) region, i.e. VH and/or VL
    • C07K2317/565Complementarity determining region [CDR]
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/70Immunoglobulins specific features characterized by effect upon binding to a cell or to an antigen
    • C07K2317/74Inducing cell proliferation
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/70Immunoglobulins specific features characterized by effect upon binding to a cell or to an antigen
    • C07K2317/75Agonist effect on antigen
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/70Immunoglobulins specific features characterized by effect upon binding to a cell or to an antigen
    • C07K2317/76Antagonist effect on antigen, e.g. neutralization or inhibition of binding
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/90Immunoglobulins specific features characterized by (pharmaco)kinetic aspects or by stability of the immunoglobulin
    • C07K2317/92Affinity (KD), association rate (Ka), dissociation rate (Kd) or EC50 value

Abstract

The present application provides isolated antibodies and antigen binding fragments thereof that specifically bind to programmed death ligand 1 (PD-L1). These PD-L1 antibodies, or antigen-binding fragments thereof, have a high affinity for PD-L1, function to inhibit PD-L1, are less immunogenic in a given species (e.g., human) than their unmodified parent antibodies, are capable of increasing T cell proliferation and IL-2 secretion in mixed lymphocyte responses, and can be used to treat human diseases (e.g., cancer, infectious disease, autoimmune disease, asthma, transplant rejection, and inflammatory disorders (inflammatory disorder)).

Description

Immunotherapy using antibodies that bind to programmed death ligand 1 (PD-L1)
Related patent application
The present application claims the benefit of U.S. provisional application No. 62/438,622, filed on date 2016, 12, 23, which is incorporated herein by reference in its entirety.
Technical Field
Immunotherapy for the treatment of cancer is rapidly evolving from therapies that globally and non-specifically mimic the immune system to more targeted activation of individual components of the immune system, leading to increased efficacy and reduced toxicity. Of primary interest are therapies that inhibit the interaction between programmed death ligand 1 (PD-L1) and programmed death 1 (PD-1), programmed death ligand 1 (PD-L1) being present on the surface of tumor or antigen presenting cells and programmed death 1 (PD-1) being present on the surface of activated lymphocytes.
Programmed death 1 (PD-1) is a member of the CD28 receptor family, which also includes CD28, CTLA-4, ICOS and BTLA. Programmed death ligand 1 (PD-L1) is one of two cell surface glycoprotein ligands for PD-1 (the other is PD-L2) that down-regulate T cell activation and cytokine secretion upon binding to PD-1 (Ohigashi et al, clin Cancer Res,11:2947-53,2005). PD-L1 has been shown to play a role in tumor immunity by increasing apoptosis of antigen-specific T cell clones (Dong et al, nat Med,8:793-800,2002). In Cancer, the PD-L1 and PD-1/PD-L1 pathways can protect tumors from cytotoxic T cells, ultimately suppressing anti-tumor immune responses by inactivating cytotoxic T cells in the tumor microenvironment, and preventing the priming and activation of new T cells in the lymph nodes and subsequent recruitment to the tumor (Chen and Irving, clin Cancer res.,18:6580-6587,2012).
Inhibition of the PD-1/PD-L1 interaction mediates potent antitumor activity in preclinical models (U.S. Pat. Nos. 8,008,449 and 7,943,743), and treatment of cancers with Ab inhibitors of the PD-1/PD-L1 interaction has entered clinical trials (see, e.g., topalian et al, curr Opin immunol.,24:207-212,2012; brahmer et al, N Engl J Med.,366 (26): 2455-65,2012; garon et al, N Engl J Med,372:2018-2028,2015; philips et al, int. Immunol.,27 (1): 39-46,2015). PD-L1 expression has been found in several murine and human cancers, including human lung, ovarian and colon cancers and multiple myelomas, and anti-PD-L1 antibodies developed by, for example, bristol-Myers Squibb (BMS-936559), medimune (MEDI 4736), genentech (MPDL 3280A), merck/Pfizer (MSB 0010718C) have been or are currently undergoing clinical evaluation.
The tolerance of PD-1 pathway blockers and their unique mechanism of action have made them ideal struts for combinatorial scheme development. Recent clinical data combining CTLA-4 and PD-L1 blockade in melanoma patients showed an increased objective tumor response rate compared to blocking either checkpoint alone, supporting the insight that combined checkpoint blockade may lead to increased clinical benefit (Wolchok et al, N Engl J Med,366:2443-54,2012).
The invention is based on the identification of novel PD-L1 antibodies and antigen-binding fragments thereof. Accordingly, the present invention relates to compositions and methods for using the novel human anti-PD-L1 antibodies and antigen-binding fragments thereof described herein for diagnosis, prognosis, and treatment of conditions that would benefit from modulation of PD-L1 and/or PD-1, such as cancer, persistent infectious disease, autoimmune disease, asthma, transplant rejection, and inflammatory disorders (inflammatory disorder).
Disclosure of the invention
In one aspect of the invention, there are provided isolated antibodies and antigen binding fragments thereof that specifically bind to programmed death ligand 1 (PD-L1). These PD-L1 antibodies, or antigen-binding fragments thereof, have a high affinity for PD-L1, function to inhibit PD-L1, are less immunogenic in a given species (e.g., human) than their unmodified parent antibodies, are capable of increasing T cell proliferation and IL-2 secretion in mixed lymphocyte responses, and can be used to treat human diseases (e.g., cancer, infectious disease, autoimmune disease, asthma, transplant rejection, and inflammatory disorders).
In various embodiments, the antibody or antigen binding fragment is selected from the group consisting of a human antibody, a humanized antibody, a chimeric antibody, a monoclonal antibody, a polyclonal antibody, a recombinant antibody, a single chain antibody, a diabody, a triabody, a tetrabody, a Fab fragment, a Fab' fragment, a Fab 2 Fragments, F (ab)' 2 Fragments, domain antibodies, igD antibodies, igE antibodies, igM antibodies, igG1 antibodies, igG2 antibodies, igG3 antibodies, igG4 antibodies, or IgG4 antibodies having at least one mutation in the hinge region that reduces the propensity to form disulfide bonds within the H chain. In various embodiments, the antibody is a chimeric antibody. In various embodiments, the antibody is a humanized antibody. In various embodiments, the antibody is a fully human antibody. In various embodiments, isolated antibodies and antigen binding fragments thereof are provided that have high affinity for the human PD-L1 protein of SEQ ID NO. 1.
In multiple casesIn one embodiment, the antibody or antigen binding fragment is present in an amount of at least about 1X 10 -6 M, at least about 1X 10 -7 M, at least about 1X 10 -8 M, at least about 1X 10 -9 M, at least about 1X 10 -10 M, at least about 1X 10 -11 M, or at least about 1X 10 -12 Dissociation constant of M (K D ) Binds to the PD-L1 protein.
In various embodiments, an isolated antibody or antigen binding fragment thereof of the invention binds to human PD-L1 and comprises any one of the following: (a) A light chain CDR3 sequence that is identical, substantially identical or substantially similar to a CDR3 sequence selected from SEQ ID NOs 10-11; (b) A heavy chain CDR3 sequence that is identical, substantially identical or substantially similar to a CDR3 sequence selected from SEQ ID NOs 5-6; or (c) a light chain CDR3 sequence of (a) and a heavy chain CDR3 sequence of (b).
In various embodiments, the isolated antibody or antigen binding fragment further comprises an amino acid sequence selected from the group consisting of: (d) A light chain CDR1 sequence that is identical, substantially identical or substantially similar to a CDR1 sequence selected from SEQ ID NOs 7-8; (e) A light chain CDR2 sequence that is identical, substantially identical or substantially similar to a CDR2 sequence selected from SEQ ID No. 9; (f) A heavy chain CDR1 sequence that is identical, substantially identical or substantially similar to a CDR1 sequence selected from SEQ ID No. 2; (g) A heavy chain CDR2 sequence that is identical, substantially identical or substantially similar to a CDR2 sequence selected from SEQ ID NOs 3-4; (h) The light chain CDR1 sequence of (d) and the heavy chain CDR1 sequence of (f); or (i) the light chain CDR2 sequence of (e) and (g) the heavy chain CDR2 sequence of (g).
In various embodiments, an isolated human monoclonal antibody or antigen binding fragment thereof of the invention binds to human PD-L1 and comprises: (a) A light chain CDR1 sequence that is identical, substantially identical or substantially similar to a CDR1 sequence selected from SEQ ID NOs 7-8; (b) A light chain CDR2 sequence that is identical, substantially identical or substantially similar to a CDR2 sequence selected from SEQ ID No. 9; (c) A light chain CDR3 sequence that is identical, substantially identical or substantially similar to a CDR3 sequence selected from SEQ ID NOs 10-11; (d) A heavy chain CDR1 sequence that is identical, substantially identical or substantially similar to a CDR1 sequence selected from SEQ ID No. 2; (e) A heavy chain CDR2 sequence that is identical, substantially identical or substantially similar to a CDR2 sequence selected from SEQ ID NOs 3-4; and (f) a heavy chain CDR3 sequence that is identical, substantially identical or substantially similar to a CDR3 sequence selected from SEQ ID NOS 5-6.
In various embodiments, an isolated human monoclonal antibody or antigen binding fragment thereof of the invention binds to human PD-L1 and comprises: (a) A light chain CDR1 sequence that is identical, substantially identical or substantially similar to SEQ ID No. 7; (b) A light chain CDR2 sequence that is identical, substantially identical or substantially similar to SEQ ID No. 9; (c) A light chain CDR3 sequence that is identical, substantially identical or substantially similar to SEQ ID No. 10; (d) A heavy chain CDR1 sequence that is identical, substantially identical or substantially similar to SEQ ID No. 2; (e) A heavy chain CDR2 sequence that is identical, substantially identical or substantially similar to SEQ ID No. 3; and (f) a heavy chain CDR3 sequence that is identical, substantially identical or substantially similar to SEQ ID NO. 5.
In various embodiments, an isolated human monoclonal antibody or antigen binding fragment thereof of the invention binds to human PD-L1 and comprises: (a) A light chain CDR1 sequence that is identical, substantially identical or substantially similar to SEQ ID No. 8; (b) A light chain CDR2 sequence that is identical, substantially identical or substantially similar to SEQ ID No. 9; (c) A light chain CDR3 sequence that is identical, substantially identical or substantially similar to SEQ ID No. 11; (d) A heavy chain CDR1 sequence that is identical, substantially identical or substantially similar to SEQ ID No. 2; (e) A heavy chain CDR2 sequence that is identical, substantially identical or substantially similar to SEQ ID No. 4; and (f) a heavy chain CDR3 sequence that is identical, substantially identical or substantially similar to SEQ ID NO. 6.
In various embodiments, the separations of the inventionBinds to human PD-L1 and comprises: (a) One or more heavy chain variable domains and/or one or more light chain variable domains, the one or more variable domains having a set of three light chain CDRs 1, CDR2, and CDR3, and/or a set of three heavy chain CDRs 1, CDR2, and CDR3, the three light chain CDRs 1, CDR2, and CDR3 being identical, substantially identical, or substantially similar to SEQ ID NOs 7-8, 9, and 10-11, the three heavy chain CDRs 1, CDR2, and CDR3 being identical, substantially identical, or substantially similar to SEQ ID NOs 2, 3-4, and 5-6; and (b) a set of four variable region framework regions from a human immunoglobulin (IgG). In various embodiments, the antibody may optionally comprise a hinge region. In various embodiments, the framework region is selected from human germline exon X H 、J H Vk and jk sequences. In various embodiments, the antibody is a fully humanized antibody. In various embodiments, the antibody is a fully human antibody.
In various embodiments, the isolated antibody or antigen binding fragment, when bound to human PD-L1: (a) Binds to human PD-L1 with a Kd that is substantially the same or greater than the reference antibody; (b) competing with the reference antibody for binding to human PD-L1; or (c) is less immunogenic than the reference antibody in a human subject, wherein the reference antibody comprises a combination of heavy chain variable domain sequences and light chain variable domain sequences selected from the group consisting of SEQ ID NOs 12/16 and 14/18.
In various embodiments, an isolated humanized antibody or antigen-binding fragment thereof of the invention binds to human PD-L1 and comprises a heavy chain variable region having a sequence that is the same, substantially the same or substantially similar to SEQ ID NOS 20, 22, 24, 26, 28, 30, 32, 34, 36, 38, 40, 42, 44 and 46 and a light chain variable region having a sequence that is the same, substantially the same or substantially similar to SEQ ID NOS 21, 23, 25, 27, 29, 31, 35, 37, 39, 41, 43, 45 and 47.
In various embodiments, an isolated humanized antibody or antigen binding fragment thereof of the invention binds to human PD-L1 and comprises the heavy chain sequence set forth in SEQ ID NO. 55 and the light chain sequence set forth in SEQ ID NO. 56.
In various embodiments, an isolated humanized antibody or antigen binding fragment thereof of the invention binds to human PD-L1 and comprises the heavy chain sequence set forth in SEQ ID NO:57 and the light chain sequence set forth in SEQ ID NO: 58.
In various embodiments, an isolated humanized antibody or antigen binding fragment thereof of the invention binds to human PD-L1 and comprises the heavy chain sequence set forth in SEQ ID NO. 59 and the light chain sequence set forth in SEQ ID NO. 60.
In various embodiments, an isolated humanized antibody or antigen binding fragment thereof of the invention binds to human PD-L1 and comprises the heavy chain sequence set forth in SEQ ID NO. 61 and the light chain sequence set forth in SEQ ID NO. 62.
In various embodiments, an isolated humanized antibody or antigen binding fragment thereof of the invention binds to human PD-L1 and comprises the heavy chain sequence set forth in SEQ ID NO. 63 and the light chain sequence set forth in SEQ ID NO. 64.
In various embodiments, the heavy chain variable region of the isolated humanized antibody of the invention binds to human PD-L1 and comprises a) FR1 selected from the group consisting of amino acids 20-49 of SEQ ID NOs 55, 57, 59, 61 and 63; b) FR2 selected from the group consisting of amino acids 55-68 of SEQ ID NO:55, 57, 59, 61 and 63; c) FR3 selected from the group consisting of amino acids 86-117 of SEQ ID NO:55, 57, 59, 61 and 63; and d) FR4 selected from the group consisting of amino acids 127-137 of SEQ ID NOS: 55, 57, 59, 61 and 63.
In various embodiments, the light chain variable region of the isolated humanized antibody of the invention binds to human PD-L1 and comprises a) FR1 selected from the group consisting of amino acids 20-42 of SEQ ID NOs 56, 58, 60, 62 and 64; b) FR2 selected from the group consisting of amino acids 54-68 of SEQ ID NO:56, 58, 60, 62 and 64; c) FR3 selected from the group consisting of amino acids 76-107 of SEQ ID NOS: 56, 58, 60, 62 and 64; and d) FR4 selected from the group consisting of amino acids 117-126 of SEQ ID NO:56, 58, 60, 62 and 64.
In another aspect, the invention relates to a pharmaceutical composition comprising an isolated antibody or antigen-binding fragment of the invention in admixture with a pharmaceutically acceptable carrier. In various embodiments, the pharmaceutical composition comprises an isolated human antibody in admixture with a pharmaceutically acceptable carrier. In various embodiments, the pharmaceutical composition is formulated for administration by a route selected from the group consisting of: subcutaneous injection, intraperitoneal injection, intramuscular injection, intrasternal injection, intravenous injection, intraarterial injection, intrathecal injection, intraventricular injection (intraventricular injection), intraurethral injection, intracranial injection, intrasynovial injection, or by infusion.
In another aspect, the invention relates to a method of treating a subject suffering from a T-cell-related disease (T-cell-related disease) or a disease that can be alleviated or prevented by boosting or suppressing or prolonging the immune response, comprising administering to the subject a therapeutically effective amount (as monotherapy or in a combination therapy regimen) of an antibody of the invention or an antigen binding fragment thereof. In various embodiments, the subject is a human subject.
In another aspect, the invention relates to a method of treating cancer in a subject, the method comprising administering to the subject a therapeutically effective amount (as monotherapy or in a combination therapy regimen) of an isolated antibody or antigen binding fragment of the invention. In various embodiments, the cancer is a cancer associated with increased expression of PD-L1. In various embodiments, the cancer is selected from the group consisting of: melanoma (e.g., metastatic malignant melanoma), colorectal cancer (CRC), renal cancer, non-small cell lung cancer (NSCLC), bladder cancer, prostate cancer, breast cancer, colon cancer, ovarian cancer, and lung cancer. In various embodiments, the subject previously responded to treatment with an anti-cancer therapy, but suffered from a relapse after cessation of the therapy (hereinafter referred to as "recurrent cancer"). In various embodiments, the subject has a resistant (resistance) cancer or refractory cancer.
In another aspect, the invention relates to a method of treating an infectious disease in a subject, the method comprising administering to the subject a therapeutically effective amount (as monotherapy or in a combination therapy regimen) of an isolated antibody or antigen binding fragment of the invention. In various embodiments, the subject has an infectious disease that is resistant to treatment with a conventional vaccine or that is not effectively treated by treatment with a conventional vaccine.
In another aspect, the invention relates to a combination therapy designed to treat cancer or an infectious disease in a subject, the combination therapy comprising administering to the subject a) a therapeutically effective amount of an isolated antibody or antigen binding fragment of the invention, and b) one or more additional therapies selected from the group consisting of immunotherapy, chemotherapy, small molecule kinase inhibitor targeted therapy, surgery, radiation therapy, vaccination regimen, and stem cell transplantation, wherein the combination therapy provides increased cell killing of tumor cells, i.e., there is a synergy between the isolated antibody or antigen binding fragment and the additional therapies when co-administered.
In another aspect, an isolated immunoconjugate or fusion protein is provided that comprises an antibody or antigen binding fragment conjugated, linked (or otherwise stably associated) with an effector molecule. In various embodiments, the effector molecule is an immunotoxin, cytokine, chemokine, therapeutic agent, or chemotherapeutic agent.
In another aspect, an antibody or antigen binding fragment disclosed herein can be covalently linked (or otherwise stably associated) with an additional functional moiety, such as a label or moiety that imparts a desired pharmacokinetic property. In various embodiments, the marker is selected from the group consisting of: fluorescent markers, radioactive markers, and markers with unique nuclear magnetic resonance characteristics.
In another aspect, the invention provides methods for detecting the presence of a human PD-L1 antigen in a sample in vitro or in vivo, e.g., methods for diagnosing a human PD-L1-related disorder.
In another aspect, an isolated nucleic acid comprising a polynucleotide sequence encoding an antibody or antigen binding fragment disclosed herein is provided. Expression vectors comprising the nucleic acids of the invention are also provided. Also provided are isolated cells comprising the expression vectors of the invention. In various embodiments, the cell is a host cell comprising an expression vector of the invention. In various embodiments, the cell is a hybridoma, wherein the chromosome of the cell comprises a nucleic acid of the invention. Also provided are methods of making an antibody or antigen-binding fragment of the invention, comprising culturing or incubating a cell under conditions that allow the cell to express the antibody or antigen-binding fragment of the invention.
Mode for carrying out the invention
The present invention relates to antigen binding proteins, such as antibodies or antigen binding fragments thereof, that specifically bind to human PD-L1. In one aspect, isolated antibodies and antigen binding fragments thereof are provided that specifically bind to PD-L1, have high affinity for PD-L1, function to inhibit PD-L1, are less immunogenic in a given species (e.g., human) than their unmodified parent antibodies, and can be used to treat human diseases (e.g., cancer), infections, and other disorders mediated by PD-L1. Nucleic acid molecules and derivatives and fragments thereof are also provided, which comprise a sequence of a polynucleotide encoding the entire polypeptide or a portion of the polypeptide that binds to PD-L1, such as a nucleic acid encoding the entire anti-PD-L1 antibody or a portion of an anti-PD-L1 antibody, an antibody fragment, or an antibody derivative. Vectors and plasmids comprising such nucleic acids, and cells or cell lines comprising such nucleic acids and/or vectors and plasmids are also provided. Also provided are methods of making, identifying, or isolating an antigen binding protein that binds to human PD-L1 (such as an anti-PD-L1 antibody), methods of determining whether an antigen binding protein binds to PD-L1, methods of making a composition (such as a pharmaceutical composition) comprising an antigen binding protein that binds to human PD-L1, and methods for administering an antibody or antigen binding fragment thereof that binds to PD-L1 to a subject, e.g., for treating a condition mediated by PD-L1.
Definition of the definition
Unless defined otherwise, scientific and technical terms used in connection with the present invention shall have the meanings commonly understood by one of ordinary skill in the art. Furthermore, unless the context requires otherwise, singular terms shall include the plural and plural terms shall include the singular. Generally, the nomenclature and techniques described herein employed in connection with cell and tissue culture, molecular biology, immunology, microbiology, genetics, and protein and nucleic acid chemistry and hybridization are those commonly employed and well known in the art. Unless otherwise indicated, the methods and techniques of the present invention are generally performed according to conventional methods well known in the art and as described in various general and more specific references cited and discussed throughout the present specification. See, e.g., green and Sambrook, molecular Cloning: A Laboratory Manual, 4 th edition, cold Spring Harbor Laboratory Press, cold Spring Harbor, n.y. (2012), which is incorporated herein by reference. Enzymatic reactions and purification techniques are performed according to the manufacturer's instructions, as commonly done in the art or as described herein. The nomenclature and the laboratory procedures and techniques described herein utilized in connection with analytical chemistry, synthetic organic chemistry, and pharmaceutical chemistry are those commonly employed and well known in the art. Standard techniques are used for chemical synthesis, chemical analysis, drug preparation, formulation and delivery, and treatment of subjects.
The polynucleotide and polypeptide sequences are indicated using standard single-letter abbreviations or three-letter abbreviations. Unless otherwise indicated, polypeptide sequences have their amino-terminal end on the left and their carboxy-terminal end on the right, and the upper strand of single-stranded and double-stranded nucleic acid sequences has their 5 '-terminal end on the left and their 3' -terminal end on the right. Specific portions of a polypeptide may be named by amino acid residue numbering, such as amino acids 80 to 119, or by the actual residues at that position, such as Ser80 to Ser 119. A particular polypeptide or polynucleotide sequence may also be described based on how it differs from a reference sequence. The polynucleotide and polypeptide sequences of a particular light chain variable domain and heavy chain variable domain are designated L1 ("light chain variable domain 1") and H1 ("heavy chain variable domain 1"). Antibodies comprising a light chain and a heavy chain are indicated by combining the name of the light chain and the name of the heavy chain variable domain. For example, "L4H7" refers to an antibody comprising, for example, a light chain variable domain of L4 and a heavy chain variable domain of H7.
The term "antibody" as used herein refers to a protein comprising one or more polypeptides, which polypeptides are substantially or partially encoded by immunoglobulin genes or fragments of immunoglobulin genes, and having specificity for a tumor antigen or for a molecule that is overexpressed in a pathological state. Putative immunoglobulin genes include kappa, lambda, alpha, gamma, delta, epsilon and mu constant region genes, as well as subtypes and a large number of immunoglobulin variable region genes of these genes. Light Chains (LC) are classified as either kappa or lambda. Heavy Chains (HC) are classified as γ, μ, α, δ or ε, which in turn define immunoglobulin classes IgG, igM, igA, igD and IgE, respectively. Typical immunoglobulin (e.g., antibody) structural units comprise tetramers. Each tetramer comprises identical two pairs of polypeptide chains, each pair having one "light chain" (about 25 kD) and one "heavy chain" (about 50-70 kD). The N-terminus of each chain defines a variable region of about 100 to 110 or more amino acids that is primarily responsible for antigen recognition.
In full length antibodies, each heavy chain comprises a heavy chain variable region (abbreviated herein as HCVR or VH) and a heavy chain constant region. The heavy chain constant region comprises 3 domains, CH1, CH2, and CH3 (and in some examples, CH 4). Each light chain comprises a light chain variable region (abbreviated herein as LCVR or VL) and a light chain constant region. The light chain constant region comprises a domain, C L . VH and VL regions can be further sub-classified as hypervariable regions known as Complementarity Determining Regions (CDRs) interspersed with regions that are more conserved, known as Framework Regions (FR). Each VH and VL comprises 3 CDRs and 4 FRs arranged from amino-terminus to carboxy-terminus in the following order: FR1, CDR1, FR2, CDR2, FR3, CDR3, FR4. The framework regions and CDR ranges have been defined. The sequences of the framework regions of the different light or heavy chains are relatively conserved in species such as humans. The framework regions of antibodies, i.e., the combined framework regions of the light and heavy chain components, are initiated in three dimensionsThe role of positioning and aligning CDRs in space. Immunoglobulin molecules may be of any type (e.g., igG, igE, igM, igD, igA and IgY), class (e.g., igG1, igG2, igG3, igG4, igA1, and IgA 2), or subclass.
CDRs are mainly responsible for binding to epitopes of antigens. The CDRs of each chain are commonly referred to as CDR1, CDR2, CDR3, numbered sequentially starting from the N-terminus, and are also commonly identified by the chain in which the particular CDR is located. Thus, VH CDR3 is located in the variable domain in which its antibody heavy chain is found, while VL CDR1 is CDR1 from the variable domain in which its antibody light chain is found. Antibodies with different specificities (i.e., different binding sites for different antigens) have different CDRs. Although CDRs vary from antibody to antibody, only a limited number of amino acid positions in the CDRs are directly involved in antigen binding. These positions in the CDRs are called Specificity Determining Residues (SDRs).
Kabat definition is a standard for numbering residues in antibodies and is commonly used to identify CDR regions. The Kabat database is now on-line and CDR sequences can be determined, see for example IMGT/V-QUEST program 3.2.18 edition, 2011, month 3, 29, and Brochet, x. Et al, nucleic acids res.36, W503-508,2008 available on the internet. The Chothia definition is similar to the Kabat definition, but the Chothia definition considers the location of certain structural loop regions. See, e.g., chothia et al, j.mol.biol.,196:901-17,1986; chothia et al Nature,342:877-83,1989.AbM defines a suite of integrated computer programs produced by Oxford Molecular Group that model antibody structures. See, e.g., martin et al, proc.Natl. Acad.Sci.USA,86:9268-9272,1989; "AbM TM A Computer Program for Modeling Variable Regions of Antibodies, "Oxford, UK; oxford Molecular, ltd. AbM definition the quaternary Structure of an antibody is modeled from a primary sequence using a combination of knowledge databases and de novo methods, such as those described by Samuldrala et al, "Ab Initio Protein Structure Prediction Using a Combined Hierarchical Approach," PROTEINS, structure, function and Genetics supply, 3:194-198,1999. The contact definition is based on analysis of the complex crystal structure available. See, e.g., macCallum Et al, J.mol.biol.,5:732-45,1996.
The term "Fc region" is used to define the C-terminal region of an immunoglobulin heavy chain that can be produced by digestion of an intact antibody with papain. The Fc region may be a native sequence Fc region or a variant Fc region. The Fc region of an immunoglobulin generally comprises two constant domains, a CH2 domain and a CH3 domain, and optionally comprises a CH4 domain. The Fc portion of an antibody mediates several important effector functions, such as cytokine induction, ADCC, phagocytosis, complement Dependent Cytotoxicity (CDC) and half-life/clearance of the antibody and antigen-antibody complex (e.g., the acidic pH of nascent FcR (FcRn) in endosomes (endosomes) binds to the Fc region of IgG and protects IgG from degradation, contributing to the long serum half-life of IgG). Replacement of amino acid residues in the Fc portion to alter antibody effector function is known in the art (see, e.g., winter et al, U.S. Pat. nos. 5,648,260 and 5,624,821).
Antibodies exist as intact immunoglobulins or as a number of well-characterized fragments. Such fragments include Fab fragments, fab' fragments, fab which bind to the target antigen 2 、F(ab)’ 2 Fragments, single chain Fv proteins ("scfvs"), and disulfide stabilized Fv proteins ("dsfvs"). scFv proteins are fusion proteins in which the light chain variable region of an immunoglobulin and the heavy chain variable region of an immunoglobulin are bound by a linker, whereas in dsFv the chains have been mutated to introduce an associated disulfide bond that stabilizes the chains. Although a variety of antibody fragments have been defined with respect to digestion of intact antibodies, the skilled artisan will appreciate that such fragments may be synthesized de novo, either chemically or by using recombinant DNA methods. Thus, as used herein, the term antibody includes, for example, monoclonal antibodies (including full length monoclonal antibodies), polyclonal antibodies, multispecific antibodies (e.g., bispecific antibodies) formed from at least 2 intact antibodies, human antibodies, humanized antibodies, camelized antibodies (camelised antibody), chimeric antibodies, single chain Fv (scFv), single chain antibodies, single domain antibodies, fab fragments, F (ab') 2 Fragments, antibody fragments exhibiting desired biological activity, disulfide-linked Fv(s)dFv), intracellular antibodies (intrabodies), and epitope-binding fragments or antigen-binding fragments of any of the above.
Papain digestion of antibodies produces two identical antigen binding fragments, known as "Fab" fragments, each with a single antigen binding site. "Fab fragment" contains the CH1 and variable regions of one light and one heavy chain. The heavy chain of a Fab molecule is unable to form disulfide bonds with another heavy chain molecule. "Fab ' fragments" comprise a light chain and a portion of a heavy chain comprising a VH domain and a CH1 domain and further comprising a region between the CH1 and CH2 domains such that an interchain disulfide bond may be formed between the two heavy chains of two Fab ' fragments to form F (ab ') 2 A molecule.
Pepsin-treated antibodies produced F (ab') with two antigen binding sites and still be able to crosslink antigens 2 Fragments. "F (ab') 2 The fragment "comprises two light chains and two heavy chains, the two heavy chains comprising a portion of the constant region between the CH1 and CH2 domains, such that an interchain disulfide bond is formed between the two heavy chains. Thus, F (ab') 2 Fragments comprise two Fab' fragments that are bound together by a disulfide bond between the two heavy chains.
The "Fv region" comprises variable regions from both the heavy and light chains, but lacks constant regions.
A "single chain antibody" is an Fv molecule in which the heavy and light chain variable regions have been joined by a flexible linker to form a single polypeptide chain that forms an antigen binding region. Single chain antibodies are discussed in detail in International patent application publication No. WO 88/01649, U.S. Pat. No. 4,946,778 and No. 5,260,203, the disclosures of which are incorporated by reference.
The terms "antigen binding fragment" and "antigen binding protein" as used herein mean any protein that binds to a particular target antigen. "antigen binding fragments" include, but are not limited to, antibodies and binding portions thereof, such as immunologically functional fragments. Exemplary antigen binding fragments of an antibody are one or more heavy chain CDRs and/or one or more light chain CDRs, or heavy chain variable regions and/or light chain variable regions.
As used herein, the term "immunologically functional fragment" (or simply "fragment") of an antibody or immunoglobulin chain (heavy or light chain) antigen binding protein is an antigen binding protein that comprises a portion of an antibody that lacks at least some of the amino acids present in the full length chain but is still capable of specifically binding to an antigen (regardless of how the portion is obtained or synthesized). Such fragments are biologically active in that they bind to the target antigen and can compete with other antigen binding proteins (including intact antibodies) for binding to a given epitope. In some embodiments, the fragment is a neutralizing fragment. In one aspect, such fragments will retain at least one CDR present in the full length light chain or heavy chain, and in some embodiments will comprise a single heavy chain and/or light chain or portion thereof. These biologically active fragments may be produced by recombinant DNA techniques or may be produced by enzymatic or chemical cleavage of antigen binding proteins, including intact antibodies. Immunologically functional immunoglobulin fragments include, but are not limited to, fab, diabodies, fab ', F (ab') 2 Fv, domain antibodies and single chain antibodies, and may be derived from any mammalian source, including but not limited to human, mouse, rat, camel, or rabbit. It is also contemplated that functional portions of the antigen binding proteins disclosed herein, such as one or more CDRs, can be covalently bound to a second protein or small molecule to produce therapeutic agents directed against specific targets in the body that have bifunctional therapeutic properties or have an extended serum half-life.
Diabodies (diabodies) are bivalent antibodies comprising two polypeptide chains, wherein each polypeptide chain comprises VH and VL regions connected by a linker that is too short to allow pairing between two regions on the same chain, thereby allowing pairing of each region with a complementary region on the other polypeptide chain (see, e.g., holliger et al, proc. Natl. Acad. Sci. USA,90:6444-48,1993; and Poljak et al, structures, 2:1121-23,1994). If the two polypeptide chains of a diabody are identical, then the diabody resulting from their pairing will have two identical antigen binding sites. Polypeptide chains having different sequences can be used to make diabodies having two different antigen binding sites. Similarly, a triabody (triabody) and a tetrabody (tetrabody) are antibodies that comprise three and four polypeptide chains, respectively, and form three and four antigen binding sites, respectively, which may be the same or different.
Bispecific antibodies or fragments can have several configurations. For example, a bispecific antibody may be similar to a single antibody (or antibody fragment) but have two different antigen binding sites (variable regions). In various embodiments, bispecific antibodies can be prepared by chemical techniques (Kranz et al, proc. Natl. Acad. Sci. USA,78:5807, 1981); by the "polyoma" technique (see, e.g., U.S. patent No. 4,474,893); or by recombinant DNA techniques. In various embodiments, bispecific antibodies of the present disclosure can have binding specificity for at least two different epitopes, at least one of which is a tumor-associated antigen. In various embodiments, the antibodies and fragments may also be xenogenous antibodies (heteroantibodies). Xenogenous antibodies are two or more antibodies or antigen binding fragments (e.g., fab) linked together, each antibody or fragment having a different specificity.
The term "monoclonal antibody" as used herein refers to an antibody obtained from a population of substantially homogeneous antibodies, i.e., the individual antibodies comprising the population are identical except for possible naturally occurring mutations that may be present in minor amounts. Monoclonal antibodies are highly specific, being directed against a single antigen. Furthermore, unlike polyclonal antibody preparations, which typically include different antibodies directed against different determinants (epitopes), each monoclonal antibody is directed against a single determinant on the antigen. The modifier "monoclonal" is not to be construed as requiring antibody production by any particular method.
The term "chimeric antibody" as used herein refers to an antibody having framework residues from one species (such as a human) and CDRs (which typically confer antigen binding) from another species, such as a murine antibody that specifically binds to the targeted antigen.
As used herein, the term "human antibody" is intended to include antibodies having variable and constant regions derived from human germline immunoglobulin sequences. The human antibodies of the present disclosure may include amino acid residues not encoded by human germline immunoglobulin sequences (e.g., mutations introduced by random mutagenesis in vitro or site-specific mutagenesis or by somatic mutation in vivo), for example in CDRs and particularly in CDR 3. However, the term "human antibody" as used herein is not intended to include antibodies in which CDR sequences derived from the germline of another mammalian species (such as a mouse) have been grafted onto human framework sequences.
The term "humanized antibody" as used herein refers to an antibody comprising humanized light chains and humanized heavy chain immunoglobulins. The humanized antibody binds to the same antigen as the donor antibody that provides the CDRs. The acceptor framework of a humanized immunoglobulin or antibody may have a limited number of substitutions through amino acids taken from the donor framework. Humanized or other monoclonal antibodies may have additional conservative amino acid substitutions that have substantially no effect on antigen binding or other immunoglobulin function. In various embodiments, the framework region is selected from human germline exon X H 、J H Vk and jk sequences. For example, for V H The humanized acceptor sequence of the FR of the domain may be selected from germline V H Exon V H 1-18 (Matsuda et al, nature Genetics 3:88-94,1993) or V H 1-2 (Shin et al, EMBO J.10:3641-3645, 1991) and for the hinge region (J) H ) Can be selected from exon J H -6 (Mattilla et al, eur. J. Immunol.25:2578-2582, 1995). In other examples, the germline V kappa exon B3 (Cox et al, eur. J. Immunol.24:827-836, 1994) and the J kappa exon J kappa-1 (Hieter et al, J. Biol. Chem.257:1516-1522, 1982) may be selected as the vectors for V L Domain humanized receptor sequences.
As used herein, the term "recombinant human antibody" is intended to include all human antibodies prepared, expressed, produced, or isolated by recombinant means, such as antibodies expressed using a recombinant expression vector transfected into a host cell; antibodies isolated from a recombinant, combinatorial human antibody library; an antibody isolated from an animal (e.g., mouse) that is transgenic for human immunoglobulin genes; or antibodies produced, expressed, produced or isolated by any other means including splicing the human immunoglobulin gene sequence to other DNA sequences. Such recombinant human antibodies have variable and constant regions derived from human germline immunoglobulin sequences. However, in various embodiments, such recombinant human antibodies undergo in vitro mutagenesis (or in vivo somatic mutagenesis when transgenic animals of human Ig sequences are used), and thus the amino acid sequences of the VH and VL regions of the recombinant antibodies are sequences that, while derived from and associated with human germline VH and VL sequences, may not naturally exist in the human antibody germline repertoire (repotoire) in vivo. All such recombinant means are well known to those of ordinary skill in the art.
The term "epitope" as used herein includes any protein determinant capable of specific binding to an immunoglobulin or T cell receptor or otherwise interacting with a molecule. Epitope determinants are generally composed of molecules (chemically active surface groupings of molecules) of chemically active surface groupings, such as amino acids or carbohydrates or sugar side chains, and generally have specific three dimensional structural characteristics as well as specific charge characteristics. Epitopes may be "linear" or "conformational". In linear epitopes, all points of interaction between a protein and an interacting molecule (such as an antibody) exist linearly along the primary amino acid sequence of the protein. In conformational epitopes, points of interaction exist across amino acid residues on the protein that are separated from each other. Once the desired epitope on the antigen is determined, it is possible to generate antibodies directed against the epitope, for example, using the techniques described in this disclosure. Alternatively, during the discovery process, the generation and characterization of antibodies may elucidate information about the desired epitope. Based on this information, it is then possible to competitively screen antibodies binding to the same epitope. One way to achieve this is to conduct cross-competition (cross-competition) studies to find antibodies that competitively bind to each other, e.g., antibodies that compete for binding to an antigen.
If the antigen-binding protein (including antibodies) is bound to a polypeptide such as by dissociation constant (K D Or the corresponding Kb, as defined below) that binds to an antigen, the antigen binding protein (including antibodies) "specifically binds" to the antigen, the dissociation constant value being at least 1 x 10 -6 M, or at least 1X 10 -7 M, or at least 1X 10 -8 M, or at least 1X 10 -9 M, or at least 1X 10 -10 M, or at least 1X 10 -11 M. An antigen binding protein that specifically binds to a human antigen of interest may also be capable of binding with the same or different affinities to the same antigen of interest from other species. The term "K" as used herein D "refers to the equilibrium dissociation constant of a particular antibody-antigen interaction.
The term "surface plasmon resonance (surface plasmon resonance)" as used herein refers to an optical phenomenon that allows for analysis of real-time biospecific interactions by detecting changes in protein concentration in a biosensor matrix, for example using BIACORE TM System (Pharmacia Biosensor AB, uppsala, sweden and Piscataway, N.J.). For further description, see Jonsson U.S. et al, ann.biol. Clin.,51:19-26,1993; jonsson U.S. et al, biotechnology, 11:620-627,1991; jonsson B.et al, J.mol.Recognit.,8:125-131,1995; and Johnsson B. Et al, anal biochem,198:268-277,1991.
The term "immunogenicity" as used herein refers to the ability of an antibody or antigen binding fragment to elicit an immune response (humoral or cellular) when administered to a recipient, and includes, for example, a human anti-mouse antibody (HAMA) response. HAMA response is initiated when T cells from a subject mount an immune response to the administered antibody. T cells then recruit B cells to produce specific "anti-antibody" antibodies.
The term "immune cell" as used herein means any cell of the hematopoietic lineage involved in modulating an immune response against an antigen (e.g., autoantigen). In various embodiments, the immune cell is, for example, a T cell, B cell, dendritic cell, monocyte, natural killer cell, macrophage, langerhans cell, or Kuffer cell.
The terms "polypeptide", "peptide" and "protein" are used interchangeably herein to refer to a polymer of amino acid residues. In various embodiments, "peptide," "polypeptide," and "protein" are chains of amino acids in which the alpha carbon of the amino acid is linked by a peptide bond. Thus the terminal amino acid at one end of the chain (amino-terminal) has a free amino group, while the terminal amino acid at the other end of the chain (carboxyl-terminal) has a free carboxyl group. As used herein, the term "amino-terminal" (abbreviated N-terminal) refers to a free α -amino group on an amino acid at the amino terminus of a peptide, or to an α -amino group of an amino acid at any other position in the peptide (imino when involved in a peptide bond). Similarly, the term "carboxy-terminal" refers to a free carboxyl group on the carboxy-terminus of a peptide, or the carboxyl group of an amino acid at any other position in the peptide. Peptides also include essentially any polyamino acid, including but not limited to peptide mimics (peptide mimetics) such as amino acids linked by ether linkages rather than amide linkages.
The term "recombinant polypeptide" as used herein is intended to include all polypeptides, including fusion molecules prepared, expressed, produced, derived or isolated by recombinant means, such as polypeptides expressed using recombinant expression vectors transfected into host cells.
Polypeptides of the disclosure include polypeptides that have been modified in any manner and for any reason, e.g., to: (1) reduced susceptibility to proteolysis, (2) reduced susceptibility to oxidation, (3) altered binding affinity for the formation of protein complexes, (4) altered binding affinity, and (5) confers or modifies other physicochemical or functional characteristics. For example, a single amino acid substitution or more than one amino acid substitution (e.g., a conservative amino acid substitution) may be made in a naturally occurring sequence (e.g., in a portion of the polypeptide other than the one or more domains that form the intermolecular contacts). "conservative amino acid substitution" refers to the substitution of an amino acid in a polypeptide with a functionally similar amino acid. The following six groups each contain amino acids that are conservative substitutions for one another:
alanine (A), serine (S) and threonine (T)
Aspartic acid (D) and glutamic acid (E)
Asparagine (N) and glutamine (Q)
Arginine (R) and lysine (K)
Isoleucine (I), leucine (L), methionine (M) and valine (V)
Phenylalanine (F), tyrosine (Y) and tryptophan (W)
"non-conservative amino acid substitutions" refer to a substitution of a member from one of these classes for a member from another class. In making such a change, according to various embodiments, the hydropathic index of amino acids may be considered (hydropathic index). Each amino acid has been assigned a hydropathic index based on the hydrophobicity and charge characteristics of the amino acid. They are: isoleucine (+4.5); valine (+4.2); leucine (+3.8); phenylalanine (+2.8); cysteine/cystine (+2.5); methionine (+1.9); alanine (+1.8); glycine (-0.4); threonine (-0.7); serine (-0.8); tryptophan (-0.9); tyrosine (-1.3); proline (-1.6); histidine (-3.2); glutamic acid (-3.5); glutamine (-3.5); aspartic acid (-3.5); asparagine (-3.5); lysine (-3.9) and arginine (-4.5).
The importance of the hydrophilic amino acid index in conferring interactive biological functions on proteins is understood in the art (see, e.g., kyte et al, 1982, J.mol. Biol. 157:105-131). It is known that certain amino acids may be substituted with other amino acids having similar hydropathic indices or scores and still retain similar biological activity. In making the change based on the hydropathic index, in various embodiments, substitutions of amino acids are included whose hydropathic index is within + -2. In various embodiments, those included within ±1, and in various embodiments, those included within ±0.5.
It is also understood in the art that similar amino acid substitutions can be made effectively based on hydrophilicity, particularly where the resulting biologically functional protein or peptide is intended for use in immunological embodiments as disclosed herein. In various embodiments, the maximum local average hydrophilicity of a protein (as determined by the hydrophilicity of its adjacent amino acids) correlates with its immunogenicity and antigenicity, i.e., with a biological property of the protein.
The following hydrophilicity values have been assigned to these amino acid residues: arginine (+3.0); lysine (+3.0); aspartic acid (+3.0.+ -. 1); glutamic acid (+3.0.+ -. 1); serine (+0.3); asparagine (+0.2); glutamine (+0.2); glycine (0); threonine (-0.4); proline (-0.5.+ -. 1); alanine (-0.5); histidine (-0.5); cysteine (-1.0); methionine (-1.3); valine (-1.5); leucine (-1.8); isoleucine (-1.8); tyrosine (-2.3); phenylalanine (-2.5) and tryptophan (-3.4). In making the change based on similar hydrophilicity values, in various embodiments, substitutions of amino acids whose hydrophilicity values are within ±2 are included, in various embodiments, those within ±1 are included, and in various embodiments, those within ±0.5 are included. Exemplary amino acid substitutions are listed in table 1.
TABLE 1
The terms "polypeptide fragment" and "truncated polypeptide" as used herein refer to a polypeptide having an amino-terminal deletion and/or a carboxy-terminal deletion as compared to the corresponding full-length protein. In various embodiments, the fragment may be, for example, at least 5, at least 10, at least 25, at least 50, at least 100, at least 150, at least 200, at least 250, at least 300, at least 350, at least 400, at least 450, at least 500, at least 600, at least 700, at least 800, at least 900, or at least 1000 amino acids in length. In various embodiments, the length of a fragment may also be, for example, up to 1000, up to 900, up to 800, up to 700, up to 600, up to 500, up to 450, up to 400, up to 350, up to 300, up to 250, up to 200, up to 150, up to 100, up to 50, up to 25, up to 10, or up to 5 amino acids. Fragments may also comprise one or more additional amino acids at either or both ends thereof, e.g., sequences of amino acids from different naturally occurring proteins (e.g., fc or leucine zipper domains) or artificial amino acid sequences (e.g., artificial linker sequences).
The terms "polypeptide variant" and "polypeptide mutant" as used herein refer to polypeptides comprising an amino acid sequence in which one or more amino acid residues are inserted into, deleted from, and/or substituted into the amino acid sequence relative to another polypeptide sequence. In various embodiments, the number of amino acid residues to be inserted, deleted or substituted can be, for example, at least 1, at least 2, at least 3, at least 4, at least 5, at least 10, at least 25, at least 50, at least 75, at least 100, at least 125, at least 150, at least 175, at least 200, at least 225, at least 250, at least 275, at least 300, at least 350, at least 400, at least 450, or at least 500 amino acids in length. Variants of the disclosure include fusion proteins.
A "derivative" of a polypeptide is a polypeptide that has been chemically modified, e.g., conjugated to another chemical moiety such as, e.g., polyethylene glycol, albumin (e.g., human serum albumin), phosphorylated, and glycosylated.
The term "% sequence identity" is used interchangeably herein with the term "% identity" and refers to the level of amino acid sequence identity between two or more peptide sequences or the level of nucleotide sequence identity between two or more nucleotide sequences when aligned using a sequence alignment program. For example, as used herein, 80% identity means the same thing as 80% sequence identity determined by a defined algorithm, and means that a given sequence is at least 80% identical to another sequence of another length. In various embodiments,% identity is selected from, for example, at least 60%, at least 65%, at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, or at least 99% or greater sequence identity to a given sequence. In various embodiments,% identity ranges from, for example, about 60% to about 70%, about 70% to about 80%, about 80% to about 85%, about 85% to about 90%, about 90% to about 95%, or about 95% to about 99%.
The term "% sequence homology" is used interchangeably herein with the term "% homology" and refers to the level of amino acid sequence homology between two or more peptide sequences or the level of nucleotide sequence homology between two or more nucleotide sequences when aligned using a sequence alignment program. For example, as used herein, 80% homology means the same thing as 80% sequence homology determined by a defined algorithm, and thus a given sequence homolog has a sequence homology of greater than 80% relative to the length of the given sequence. In various embodiments,% homology is selected from, for example, at least 60%, at least 65%, at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, or at least 99% or greater sequence homology to a given sequence. In various embodiments,% homology is in the range of, for example, about 60% to about 70%, about 70% to about 80%, about 80% to about 85%, about 85% to about 90%, about 90% to about 95%, or about 95% to about 99%.
Exemplary computer programs that can be used to determine identity between two sequences include, but are not limited to, suites of BLAST programs, such as BLASTN, BLASTX and TBLASTX, BLASTP and TBLASTN, publicly available on the internet on the NCBI's website. See also Altschul et al, J.mol. Biol.215:403-10,1990 (see especially the default settings disclosed, i.e. parameters w=4, t=17) and Altschul et al, nucleic Acids Res.,25:3389-3402,1997. When evaluating a given amino acid sequence relative to the amino acid sequences in GenBank protein sequences and other public databases, sequence searches are typically performed using BLASTP programs. The BLASTX program is preferably used to search for nucleic acid sequences that have been translated in all reading frames against amino acid sequences in GenBank protein sequences and other public databases. Both BLASTP and BLASTX were run using the BLOSUM-62 matrix with a default parameter of an open gap (gap) penalty of 11.0 and an extended gap penalty of 1.0. See above.
In addition to calculating percent sequence identity, the BLAST algorithm also performs statistical analysis of the similarity between two sequences (see, e.g., karlin & Altschul, proc. Nat' l. Acad. Sci. USA,90:5873-5787,1993). One measure of similarity provided by the BLAST algorithm is the minimum total probability (P (N)), which provides an indication of the probability that a match between two nucleotide or amino acid sequences will occur by chance. For example, a nucleic acid is considered similar to a reference sequence if the smallest sum probability in a comparison of the test nucleic acid to the reference nucleic acid is, for example, less than about 0.1, less than about 0.01, or less than about 0.001.
In the context of polypeptide sequences, the term "substantially similar (substantial similarity)" or "substantially similar (substantially similar)" indicates that a polypeptide region has a sequence that is at least 70%, typically at least 80%, more typically at least 85%, or at least 90% or at least 95% sequence similarity to a reference sequence. For example, a polypeptide is substantially similar to a second polypeptide, e.g., wherein the two peptides differ by one or more conservative substitutions.
"Polynucleotide" refers to a polymer comprising nucleotide units. Polynucleotides include naturally occurring nucleic acids, such as deoxyribonucleic acid ("DNA") and ribonucleic acid ("RNA"), as well as nucleic acid analogs. Nucleic acid analogs include analogs that comprise a non-naturally occurring base, a nucleotide that is joined to other nucleotides (engage) with a linkage other than a naturally occurring phosphodiester linkage, or a nucleotide that comprises a base attached by a linkage other than a phosphodiester linkage. Thus, nucleotide analogs include, for example and without limitation, phosphorothioates, phosphorodithioates, phosphotriesters (phosphotriesters), phosphoramidates (phosphoramidates), borophosphoates (borophosphophosphates), methylphosphonates, chiral methylphosphonates, 2-O-methyl ribonucleotides, peptide Nucleic Acids (PNAs), and the like. Such polynucleotides may be synthesized, for example, using an automated DNA synthesizer. The term "nucleic acid" generally refers to a large polynucleotide. The term "oligonucleotide" generally refers to short polynucleotides, typically no more than about 50 nucleotides. It will be appreciated that when the nucleotide sequence is represented by a DNA sequence (i.e., A, T, G, C), this also includes RNA sequences in which "U" replaces "T" (i.e., A, U, G, C).
The polynucleotide sequences are described herein using conventional symbols: the left hand end of the single stranded polynucleotide sequence is the 5' -end; the left hand direction of the double stranded polynucleotide sequence is referred to as the 5' -direction. The 5 'to 3' direction of nucleotide addition to nascent RNA transcripts is referred to as the transcription direction. DNA strands having the same sequence as mRNA are referred to as "coding strands"; the sequence 5 'on the DNA strand having the same sequence as the mRNA transcribed from the DNA and located at the 5' -end of the RNA transcript is referred to as the "upstream sequence"; the sequence that is 3 'on the DNA strand having the same sequence as the RNA and that encodes the 3' -end of the RNA transcript is referred to as the "downstream sequence".
"complementary" refers to the topological compatibility or matching of the interacting surfaces of two polynucleotides. Thus, the two molecules may be described as complementary, and further, the contact surface features are complementary to each other. A first polynucleotide is complementary to a second polynucleotide if the nucleotide sequence of the first polynucleotide is substantially identical to the nucleotide sequence of the polynucleotide binding partner of the second polynucleotide, or if the first polynucleotide can hybridize to the second polynucleotide under stringent hybridization conditions.
"specifically hybridizes" or "selectively hybridizes" refers to the binding, duplexing, or hybridizing of a nucleic acid molecule to a particular nucleotide sequence under stringent conditions when the sequence is present in a complex mixture (e.g., total cell) DNA or RNA. The term "stringent conditions" refers to conditions under which a probe will preferentially hybridize to its target sequence, and to a lesser extent to other sequences, or not to other sequences at all. In the context of nucleic acid hybridization experiments such as DNA hybridization and RNA hybridization, "stringent hybridization" and "stringent hybridization wash conditions" are sequence-dependent and are different under different environmental parameters. Extensive guidance for hybridization of nucleic acids can be found in the following: tijssen,1993,Laboratory Techniques in Biochemistry and Molecular Biology-Hybridization with Nucleic Acid Probes, section I, chapter 2, "Overview of principles of hybridization and the strategy of nucleic acid probe assays", elsevier, n.y.; sambrook et al, 2001,Molecular Cloning:A Laboratory Manual,Cold Spring Harbor Laboratory,3.sup.rd.ed,NY; and Ausubel et al, current edition, current Protocols in Molecular Biology, greene Publishing Associates and Wiley Interscience, N.Y..
Generally, highly stringent hybridization and wash conditions are selected to be about 5 ℃ below the thermal melting point (Tm) for a particular sequence at a defined ionic strength and pH. Tm is the temperature (at defined ionic strength and pH) at which 50% of the target sequence hybridizes to a perfectly matched probe. Very stringent conditions are selected to be equal to the Tm of the particular probe. On filters in southern or northern blots, an example of stringent hybridization conditions for hybridization of complementary nucleic acids having more than about 100 complementary residues is 50% formalin with 1mg heparin at 42 ℃, wherein hybridization is performed overnight. An example of highly stringent wash conditions is 0.15M NaCl at 72℃for about 15 minutes. An example of stringent wash conditions is a 0.2 XSSC wash at 65℃for 15 minutes. For a description of SSC buffers, see Sambrook et al. The high stringency wash may be preceded by a low stringency wash to remove background probe signal. An exemplary medium stringency wash for a duplex of, for example, more than about 100 nucleotides is at 45 ℃,1 x SSC, for 15 minutes. An exemplary low stringency wash for a duplex of, for example, more than about 100 nucleotides is at 40 ℃,4-6 x SSC, for 15 minutes. In general, a signal to noise ratio of 2× (or higher) than that observed for unrelated probes in a particular hybridization assay indicates detection of specific hybridization.
"primer" refers to a polynucleotide that is capable of specifically hybridizing to a designated polynucleotide template and providing a point of initiation for synthesis of a complementary polynucleotide. Such synthesis occurs when the polynucleotide primer is placed under conditions in which synthesis is induced, i.e., in the presence of nucleotides, a complementary polynucleotide template, and an agent for polymerization, such as a DNA polymerase. The primer is typically single stranded, but may be double stranded. Primers are typically deoxyribonucleic acids, but a wide variety of synthetic and naturally occurring primers are useful for many applications. The primer is complementary to the template, which is designed to hybridize to the template to serve as a site for initiation of synthesis, but need not reflect the exact sequence of the template. In such cases, the specific hybridization of the primer to the template depends on the stringency of the hybridization conditions. The primer may be labeled with, for example, a chromogenic, radioactive or fluorescent moiety and used as a detectable moiety.
When used in reference to a polynucleotide, a "probe" refers to a polynucleotide that is capable of specifically hybridizing to a designated other polynucleotide sequence. The probe hybridizes specifically to the target complementary polynucleotide, but need not reflect the exact complementary sequence of the template. In such cases, the specific hybridization of the probe to the target depends on the stringency of the hybridization conditions. The probes may be labeled with, for example, chromogenic, radioactive or fluorescent moieties and used as detectable moieties. In examples where the probe provides a starting point for synthesis of the complementary polynucleotide, the probe may also be a primer.
A "vector" is a polynucleotide that can be used to introduce another nucleic acid linked thereto into a cell. One type of vector is a "plasmid," which refers to a linear or circular double-stranded DNA molecule into which additional nucleic acid segments may be ligated. Another type of vector is a viral vector (e.g., replication defective retroviruses, adenoviruses, and adeno-associated viruses), wherein additional DNA segments may be introduced into the viral genome. Some vectors are capable of autonomous replication in a host cell into which they are introduced (e.g., bacterial vectors comprising a bacterial origin of replication and episomal mammalian vectors (episomal mammalian vector)). Other vectors (e.g., non-episomal mammalian vectors) are integrated into the genome of a host cell upon introduction into the host cell, and thereby are replicated along with the host genome. An "expression vector" is a type of vector that can direct the expression of a selected polynucleotide.
A "regulatory sequence" is a nucleic acid that affects the expression (e.g., level, timing, or location of expression) of a nucleic acid to which it is operably linked. The regulatory sequence may, for example, exert its effect directly on the nucleic acid being regulated, or by the effect of one or more other molecules (e.g., polypeptides that bind to the regulatory sequence and/or nucleic acid). Examples of regulatory sequences include promoters, enhancers and other expression control elements (e.g., polyadenylation signals). Other examples of regulatory sequences are described, for example, in Goeddel,1990,Gene Expression Technology:Methods in Enzymology 185,Academic Press,San Diego,Calif, and Baron et al, 1995,Nucleic Acids Res.23:3605-06. A nucleotide sequence is "operably linked" to a regulatory sequence if the regulatory sequence affects the expression (e.g., the level, timing, or location of expression) of the nucleotide sequence.
A "host cell" is a cell that can be used to express a polynucleotide of the present disclosure. The host cell may be a prokaryote, such as e.coli (e.coli), or the host cell may be a eukaryote, such as a single cell eukaryote (e.g., yeast or other fungi), a plant cell (e.g., tobacco or tomato plant cells), an animal cell (e.g., a human cell, monkey cell, hamster cell, rat cell, mouse cell, or insect cell), or a hybridoma. Typically, a host cell is a cultured cell that can be transformed or transfected with a nucleic acid encoding a polypeptide, which can then be expressed in the host cell. The phrase "recombinant host cell" may be used to refer to a host cell that has been transformed or transfected with a nucleic acid to be expressed. The host cell may also be a cell that comprises a nucleic acid but does not express the nucleic acid at a desired level unless a regulatory sequence is introduced into the host cell such that the regulatory sequence becomes operably linked to the nucleic acid. It will be understood that the term host cell refers not only to a particular subject cell, but also to the progeny or potential progeny of such a cell. Because certain modifications may occur in succeeding generations due to, for example, mutation or environmental influences, such progeny may not, in fact, be identical to the parent cell, but are still included within the scope of the term as used herein.
The term "isolated molecule" (wherein the molecule is, for example, a polypeptide or polynucleotide) is such a molecule: the molecule is (1) not associated with its naturally associated components that accompany it in its natural state by virtue of its origin or derivation, (2) substantially free of other molecules from the same species, (3) expressed by cells from a different species, or (4) not found in nature. Thus, a molecule that is chemically synthesized, or expressed in a cell system that is different from the cell from which it naturally originates, will be "isolated" from its naturally associated components. Purification techniques well known in the art can also be used to render the molecule substantially free of naturally associated components by isolation. Molecular purity or homogeneity can be determined by a variety of means well known in the art. For example, the purity of a polypeptide sample can be determined using polyacrylamide gel electrophoresis and staining of the gel to visualize the polypeptide using techniques well known in the art. For some purposes, higher resolution may be provided by using HPLC or other means for purification well known in the art.
A protein or polypeptide is "substantially pure," "substantially homogenous," or "substantially purified" when at least about 60% to 75% of the sample appears as a single species of polypeptide. The polypeptide or protein may be monomeric or multimeric (multimeric). A substantially pure polypeptide or protein will typically comprise about 50%, 60%, 70%, 80% or 90% W/W protein sample, more typically about 95%, and preferably will be more than 99% pure. Protein purity or homogeneity can be indicated by a number of means well known in the art, such as polyacrylamide gel electrophoresis of a protein sample, followed by visualization of individual polypeptide bands after staining the gel with a stain well known in the art. For some purposes, higher resolution may be provided by using HPLC or other means for purification well known in the art.
"linker" refers to a molecule that links two other molecules, either covalently or through ionic, van der Waals, or hydrogen bonding, for example, a nucleic acid molecule that hybridizes to one complementary sequence at the 5 'end and to another complementary sequence at the 3' end, thus linking two non-complementary sequences. "cleavable linker" refers to a linker that can be degraded or otherwise cleaved to separate two components connected by the cleavable linker. Cleavable linkers are typically cleaved by enzymes, typically peptidases, proteases, nucleases, lipases, and the like. Cleavable linkers may also be cleaved by environmental factors such as, for example, changes in temperature, pH, salt concentration, etc.
The term "label" or "labeled" as used herein refers to the incorporation of another molecule in an antibody. In one embodiment, the label is a detectable label, such as a polypeptide incorporating a radiolabeled amino acid or attached to a biotin-based moiety that can be detected by a labeled avidin (e.g., streptavidin comprising a fluorescent label or enzymatic activity that can be detected by optical methods or calorimetry). In another embodiment, the label or marker may be therapeutic, such as a drug conjugate or toxin. Various methods of labeling polypeptides and glycoproteins are known in the art and may be used. Examples of markers for polypeptides include, but are not limited to, the following: radioisotopes or radionuclides (e.g 3 H、 14 C、 15 N、 35 S、 90 Y、 99 Tc、 111 In、 125 I、 131 I) The method comprises the steps of carrying out a first treatment on the surface of the Fluorescent labels (e.g., FITC, rhodamine, lanthanide fluorophores); enzyme labels (e.g., horseradish peroxidase, beta-galactosidase, luciferase, alkaline phosphatase); a chemiluminescent marker; a biotin-based group; a predetermined polypeptide epitope (e.g., leucine zipper pairing sequence, binding site of a second antibody, metal binding domain, epitope tag) recognized by a second reporter; magnetic agents (magnetic agents), such as gadolinium chelates; toxins such as pertussis toxin (pertussis toxin), taxol, cytochalasin B (cyt)The pharmaceutical compositions may include, but are not limited to, ohalasin B), gramicidin D (gramicidin D), ethidium bromide (ethidium bromide), ipecine (emetine), mitomycin (mitomycin), etoposide (etoposide), teniposide (teniposide), vincristine (vincristine), vinblastine (vinblastine), colchicine (colchicine), doxorubicin (doxorubiin), daunorubicin (daunorubicin), dihydroxyanthracene dione (dihydroxy anthracin dione), mitoxantrone (mitoxantrone), mithramycin (mithramycin), actinomycin D (actinomycin D), 1-dehydrotestosterone (1-dehydrotestosterone), glucocorticoid (glucocorticoids), procaine (procaine), tetracaine (tetracaine), lidocaine (lidocaine), propranol (procyanine), and puromycin (puromycin), and analogues thereof. In some embodiments, the tag is attached by spacer arms (spacer arms) of various lengths to reduce potential steric hindrance.
As used herein, the term "immunotherapy" refers to cancer treatment, including but not limited to treatment with depleting antibodies (depleting antibody) against specific tumor antigens; treatment with antibody-drug conjugates; treatment with agonistic, antagonistic or blocking antibodies against co-stimulatory or co-inhibitory molecules (immune checkpoints) such as PD-1, PD-L1, OX-40, CD137, GITR, LAG3, TIM-3 and VISTA; use of bispecific T cell engagement antibodiesTreatment such as blepharomab (blinatumomab); treatment involving administration of biological response modifiers (modifiers) such as IL-2, IL-12, IL-15, IL-21, GM-CSF, IFN- α, IFN- β, and IFN- γ; treatment with therapeutic vaccines such as sipuleucel-T; treatment with dendritic cell vaccines or tumor antigen peptide vaccines; treatment using Chimeric Antigen Receptor (CAR) -T cells; treatment with CAR-NK cells; treatment with Tumor Infiltrating Lymphocytes (TILs); treatment with adoptively transferred anti-tumor T cells (ex vivo expanded and/or TCR transgenic); treatment with TALL-104 cells; and the use of immunostimulants such as Toll-like receptor (TLR) agonists CpG and imiquimod Is a therapeutic agent.
The term "immunoconjugate" or "fusion protein" as used herein refers to a molecule comprising an antibody or antigen-binding fragment thereof conjugated (or linked) directly or indirectly to an effector molecule. The effector molecule may be a detectable label, an immunotoxin, a cytokine, a chemokine, a therapeutic agent, or a chemotherapeutic agent. The antibody or antigen binding fragment thereof may be conjugated to the effector molecule via a peptide linker. The immunoconjugate and/or fusion protein retains the immunoreactivity of the antibody or antigen-binding fragment, e.g., the antibody or antigen-binding fragment has approximately the same or only slightly reduced ability to bind antigen after conjugation as before conjugation. As used herein, an immunoconjugate may also be referred to as an Antibody Drug Conjugate (ADC). Because immunoconjugates and/or fusion proteins were originally prepared from two molecules with separate functions, such as antibodies and effector molecules, they are sometimes also referred to as "chimeric molecules.
"pharmaceutical composition" refers to a composition suitable for pharmaceutical use in an animal. The pharmaceutical composition comprises a pharmacologically effective amount of the active agent and a pharmaceutically acceptable carrier. "pharmacologically effective amount" refers to an amount of an agent that is effective to produce the desired pharmacological result. By "pharmaceutically acceptable carrier" is meant any standard pharmaceutical carrier, vehicle, buffer and excipient, such as phosphate buffered saline solution, aqueous 5% dextrose solution, and emulsions, such as oil/water or water/oil emulsions, and various types of wetting agents and/or adjuvants. Suitable pharmaceutical carriers and formulations are described in Remington, pharmaceutical Sciences, 21 st edition 2005,Mack Publishing Co,Easton. "pharmaceutically acceptable salts" refers to salts of compounds that may be formulated for pharmaceutical use, including, for example, salts of metals (sodium, potassium, magnesium, calcium, etc.) and salts of ammonia or salts of organic amines.
The terms "treatment", "treatment" and "treatment" refer to a method of alleviating or eliminating a biological disorder and/or at least one concomitant symptom thereof. As used herein, "alleviating" a disease, disorder, or condition means reducing the severity and/or frequency of symptoms of the disease, disorder, or condition. As used herein, "treatment" is a method for achieving a beneficial or desired clinical result. For the purposes of the present invention, beneficial or desired clinical results include, but are not limited to, any one or more of the following: alleviating one or more symptoms, alleviating the extent of a disease, preventing or delaying the spread of a disease (e.g., metastasis, such as to the lung or to lymph nodes), preventing or delaying the recurrence of a disease, delaying or slowing the progression of a disease, ameliorating the disease state, and alleviating (whether partial or total). "treatment" also includes reducing the pathological consequences of a proliferative disease. The methods of the invention contemplate any one or more of these aspects of treatment.
The term "effective amount" or "therapeutically effective amount" as used herein refers to an amount of a compound or composition that is sufficient to treat a particular disorder, condition, or disease, such as to ameliorate, alleviate, mitigate, and/or delay one or more symptoms thereof. In referring to NHL and other cancers or other unwanted cell proliferation, an effective amount comprises an amount sufficient to (i) reduce the number of cancer cells; (ii) reducing tumor size; (iii) Inhibit, delay, slow and preferably stop cancer cell infiltration into surrounding organs to some extent; (iv) Inhibit (i.e., slow to some extent, and preferably stop) tumor metastasis; (v) inhibiting tumor growth; (vi) preventing or delaying the onset and/or recurrence of a tumor; and/or (vii) an amount that alleviates to some extent one or more symptoms associated with the cancer. The effective amount may be administered in one or more administrations.
"resistant or refractory cancer" refers to tumor cells or cancers that do not respond to prior anti-cancer therapies, including, for example, chemotherapy, surgery, radiation therapy, stem cell transplantation, and immunotherapy. Tumor cells may be resistant or refractory at the beginning of treatment, or they may become resistant or refractory during treatment. Refractory tumor cells include tumors that do not respond at the beginning of treatment, or tumors that initially respond for a short period of time but do not respond to treatment. Refractory tumor cells also include tumors that respond to treatment with an anti-cancer therapy but cannot respond to subsequent rounds of therapy. Refractory tumor cells for the purposes of the present invention also include tumors that appear to be inhibited by treatment with an anti-cancer therapy but recur up to 5 years, sometimes up to 10 years or more, after cessation of treatment. The anti-cancer therapy may use chemotherapeutic agents alone, radiation alone, targeted therapy alone, surgery alone, or a combination thereof. For ease of description, and not limitation, it will be understood that refractory tumor cells are interchangeable with resistant tumors.
It will be understood that the aspects and embodiments of the invention described herein include "consisting of" and/or "consisting essentially of" the described aspects and embodiments.
Herein, reference to "about" a value or parameter includes (and describes) a variation for that value or parameter itself. For example, a description referring to "about X" includes a description of "X".
As used herein and in the appended claims, the singular forms "a," "an," "the," and "the" include plural referents unless the context clearly dictates otherwise. It will be understood that aspects and variations of the invention described herein include "consisting of" and/or "consisting essentially of" the described aspects and variations.
PD-L1 antigen
The term "PD-L1" as used herein includes variants, isoforms and species homologs of human PD-L1 (hPD-L1), hPD-L1 and analogs having at least one common epitope with hPD-L1. In various embodiments, a hPD-L1 polypeptide as used herein may comprise the amino acid sequence set forth in NCBI reference sequence NP-054862.1 (SEQ ID NO: 1):
in various embodiments, the PD-L1 polypeptide comprises an amino acid sequence that shares, for example, at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, or at least 99% homology (observed homology) with the human PD-L1 sequence of SEQ ID NO. 1. In some embodiments, the PD-L1 polypeptide has an activity of at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, at least 1×, at least 1.5×, at least 2×, at least 2.5×, or at least 3× of human PD-L1 of SEQ ID No. 1. In this context, polypeptide variants of PD-L1 can be described by reference to additions, deletions or substitutions of amino acid residues present at a given position in the sequence of 233 amino acids of SEQ ID NO. 1. Thus, for example, the term "P21W" refers to the fact that the "P" (proline, in the standard single letter code) residue at position 21 of SEQ ID NO:1 has been replaced with "W" (tryptophan, in the standard single letter code).
Antibodies to
Methods for generating novel antibodies that bind to human PD-L1 polypeptides are known to those of ordinary skill in the art. For example, a method for generating a monoclonal antibody that specifically binds to a PD-L1 polypeptide can include administering to a mouse an amount of an immunogenic composition comprising a PD-L1 polypeptide effective to stimulate a detectable immune response, obtaining antibody-producing cells (e.g., cells from the spleen) from the mouse and fusing the antibody-producing cells with myeloma cells to obtain an antibody-producing hybridoma, and testing the antibody-producing hybridoma to identify a hybridoma that produces a monoclonal antibody that specifically binds to the PD-L1 polypeptide. After obtaining, the hybridomas may be propagated in cell culture, optionally in culture conditions in which cells derived from the hybridomas produce monoclonal antibodies that specifically bind to the PD-L1 polypeptide. Monoclonal antibodies can be purified from cell cultures. A variety of different techniques are then available for testing antibodies for antigen interactions to identify particularly desirable antibodies.
Other suitable methods of producing or isolating antibodies with the requisite specificity may be used, including, for example, methods of selecting recombinant antibodies from libraries, or methods that rely on immunization of transgenic animals (e.g., mice) capable of producing a complete reservoir of human antibodies. See, e.g., jakobovits et al, proc.Natl. Acad.Sci.USA,90:2551-2555,1993; jakobovits et al Nature,362:255-258,1993; lonberg et al, U.S. Pat. nos. 5,545,806; surani et al, U.S. patent No. 5,545,807.
Antibodies can be engineered in a variety of ways. They can be prepared as single chain antibodies (including small modular immunopharmaceuticals (small modular immunopharmaceutical) or SMIPs) TM ) Fab and F (ab') 2 Fragments, etc. Antibodies may be humanized, chimeric, deimmunized or fully human. Numerous publications list many types of antibodies and methods of engineering such antibodies. See, for example, U.S. patent No. 6,355,245; 6,180,370; no. 5,693,762; 6,407,213; 6,548,640; 5,565,332; 5,225,539; 6,103,889; and No. 5,260,203.
Chimeric antibodies may be produced by recombinant DNA techniques known in the art. For example, the gene encoding the Fc constant region of a murine (or other species) monoclonal antibody molecule is digested with a restriction enzyme to remove the region encoding murine Fc and replaced with an equivalent portion of the gene encoding the human Fc constant region (see Robinson et al, international patent publication PCT/US86/02269; akira, et al, european patent application 184,187; taniguchi, M., european patent application 171,496; morrison et al, european patent application 173,494; neuberger et al, international application WO 86/01533; capilli et al, U.S. Pat. No. 4,816,567; cabilly et al, european patent application 125,023; better et al, science,240:1041-1043,1988; liu et al, PNAS USA,84:3439-3443,1987; liu et al, J. Immunol. 139:3521-35AS, 1987; PNA, 84-218, 1987; shadow, and Prinser, 35-35, 1987; naja, U.S. Pat. No. 4,816, U.S. Pat. 4,567; caster et al, european patent application 125,023; better et al, pr. Prinser.35, 1987; prinser.35, and Prinsepia, 35-35, 1987, and Nat.35.37).
Methods for humanizing antibodies have been described in the art. In practice, humanized antibodies are typically human antibodies in which some hypervariable region residues and possibly some framework region residues are replaced with residues from similar sites in rodent antibodies. Thus, such "humanized" antibodies are chimeric antibodies in which substantially less than one complete human variable region has been replaced with a corresponding sequence from a non-human species. To some extent, this can be achieved in connection with humanization techniques and display techniques using appropriate libraries. It will be appreciated that murine antibodies or antibodies from other species may be humanized or primatized using techniques well known in the art (see, e.g., winter et al, immunol Today,14:43-46,1993; and Wright et al, crit. Reviews in Immunol.,12125-168, 1992). Antibodies of interest may be engineered by recombinant DNA techniques to replace CH1, CH2, CH3, hinge domains and/or framework domains with corresponding human sequences (see WO 92/02190 and U.S. Pat. nos. 5,530,101, 5,585,089, 5,693,761, 5,693,792, 5,714,350 and 5,777,085). Furthermore, the use of Ig cDNA for the construction of chimeric immunoglobulin genes is known in the art (Liu et al, P.N.A.S.84:3439,1987; J.Immunol.139:3521, 1987). mRNA is isolated from hybridomas or other antibody-producing cells and used to produce cDNA. The cDNA of interest can be amplified by polymerase chain reaction using specific primers (U.S. Pat. Nos. 4,683,195 and 4,683,202). Alternatively, libraries are prepared and screened to isolate sequences of interest. The DNA sequence encoding the variable region of the antibody is then fused to a human constant region sequence. The sequence of the human constant region gene can be found in Kabat et al (1991) Sequences of Proteins of Immunological Interest, N.I.H. publication No. 91-3242. Human C region genes are readily available from known clones. The selection of isoforms will be guided by the desired effector functions such as complement fixation (complement fixation) or activity of antibody dependent cytotoxicity. In various embodiments, the isotype is selected from the group consisting of IgG1, igG2, igG3, and IgG 4. Either human light chain constant region kappa or lambda may be used. The chimeric, humanized antibody is then expressed by conventional methods.
U.S. Pat. No. 5,693,761 to Queen et al discloses improvements to Winter et al for humanizing antibodies, and based on the premise that: attributing affinity (avidity) loss to the humanized frameworkStructural motif problems, due to steric or other chemical incompatibilities, interfere with CDR folding into a conformation that is capable of binding found in mouse antibodies. To solve this problem, queen teaches the use of a human framework sequence that is closely homologous to the framework sequence of the mouse antibody to be humanized. Thus, the method of Queen focuses on comparing framework sequences between species. Typically, all available human variable region sequences are compared to a particular mouse sequence and percent identity between the corresponding framework residues is calculated. The human variable region with the highest percentage is selected to provide the framework sequence for the humanised item. Queen also teaches that it is important to retain certain amino acid residues from the mouse framework in the humanized framework that are critical for supporting the CDRs in a conformation that is capable of binding. The potential importance is assessed from a molecular model. Candidate residues for retention are typically adjacent to the CDR in the linear sequence or physically at any CDR residue Those within.
In other methods, once a low affinity humanized construct is obtained, the importance of a particular framework amino acid residue is determined experimentally by recovering (version) individual residues to the mouse sequence and determining antigen binding, as described by Riechmann et al, 1988. Another exemplary method for identifying important amino acids in a framework sequence is disclosed by Carter et al, U.S. Pat. No. 5,821,337 and Adair et al, U.S. Pat. No. 5,859,205. These references disclose specific Kabat residue positions in the framework that may require substitution with corresponding mouse amino acids in the humanized antibody to maintain avidity.
Another method of humanizing antibodies, known as "frame shuffling (framework shuffling)", relies on the generation of combinatorial libraries having non-human CDR variable regions fused in-frame (in frames) into pools of individual human germline frameworks (Dall' Acqua et al Methods,36:43, 2005). The library is then screened to identify clones encoding humanized antibodies that retain good binding.
The choice of the human variable regions (both light and heavy chains) to be used in the preparation of the desired humanized antibodies is very important for reducing antigenicity. The sequence of the variable region of a rodent antibody is screened against an entire library of known human variable domain sequences according to a method known as "best fit". Then, the human sequence closest to the rodent sequence was accepted as the human framework region (framework region) for the humanized antibody (Sims et al, j.immunol.,151:2296,1993; chothia et al, j.mol. Biol.,196:901, 1987). Another approach uses specific framework regions derived from the consensus sequences of all human antibodies of a specific subgroup of light chain variable regions or heavy chain variable regions. The same framework can be used for several different humanized antibodies (Carter et al, proc. Natl. Acad. Sci. USA,89:4285,1992; presta et al, J. Immunol.,151:2623, 1993).
The choice of non-human residues substituted into the human variable region may be affected by a variety of factors. These factors include, for example, the rarity of amino acids in a particular position, the likelihood of interaction with CDRs or antigens, and the likelihood of participation in the interface between the light chain variable domain interface and the heavy chain variable domain interface. (see, e.g., U.S. Pat. nos. 5,693,761, 6,632,927, and 6,639,055). One way to analyze these factors is by using a three-dimensional model of the non-human sequence and the humanized sequence. Three-dimensional immunoglobulin models are generally available and familiar to those skilled in the art. Computer programs are available that illustrate and display the possible three-dimensional conformational structures of selected candidate immunoglobulin sequences. Examination of these displays allows analysis of the possible role of residues in the function of the candidate immunoglobulin sequence, e.g., analysis of residues that affect the ability of the candidate immunoglobulin to bind to its antigen. In this manner, non-human residues may be selected and substituted for human variable region residues in order to achieve desired antibody characteristics, such as increased affinity for one or more target antigens.
Methods for preparing fully human antibodies have been described in the art. By way of example, a method for producing an anti-PD-L1 antibody or antigen-binding fragment thereof comprises the steps of: synthesizing a library of human antibodies on phage, screening the library with PD-L1 or an antibody-binding portion thereof, isolating phage that bind to PD-L1, and obtaining antibodies from phage. By way of another example, one method for preparing a library of antibodies for use in phage display technology comprises the steps of: immunization of a non-human animal comprising a human immunoglobulin locus with PD-L1 or an antigenic portion thereof to generate an immune response, extracting antibody-producing cells from the immunized animal, isolating RNA encoding the heavy and light chains of the antibodies of the invention from the extracted cells, reverse transcribing the RNA to generate cDNA, amplifying the cDNA using primers, and inserting the cDNA into a phage display vector such that the antibodies are expressed on phage. The recombinant anti-PD-L1 antibodies of the invention can be obtained in this manner.
The recombinant human anti-PD-L1 antibodies of the invention can also be isolated by screening a recombinant combinatorial antibody library. Preferably, the library is a scFv phage display library generated using human VL and VH cdnas prepared from mRNA isolated from B cells. Methods for preparing and screening such libraries are known in the art. Kits for generating phage display libraries are commercially available (e.g., pharmacia recombinant phage antibody system, catalog number 27-9400-01; and Stratagene SurfZAP) TM Phage display kit, catalog no 240612). There are other methods and reagents that can be used in the generation and screening of antibody display libraries (see, e.g., U.S. Pat. nos. 5,223,409; PCT publication Nos. WO 92/18619, WO 91/17271, WO 92/20791, WO 92/15679, WO 93/01188, WO 92/01047, and WO 92/09690; fuchs et al, bio/Technology 9:1370-1372 (1991), hay et al, hum. Anti.hybrid 3:81-85,1992, huse et al, science 246:1275-1281,1989, mcCafferty et al, nature 348:552-554,1990, griffis et al, EMBO J.12:725-734,1993, hawkins et al, J.mol.biol.226:889-896,1992, clackson et al, nature 352:624-628,1991, gram et al, proc.Natl.Acad.Sci.USA 89:3576-3580,1992, garrad et al, bio/Technology 9:1373-1377,1991, hoogeboom et al, nuc.Acid 4119-37, 1991, and Proc.Nature et al.USA 88:7978-7982,1991, each of which is incorporated herein by reference for the purpose of teaching the preparation and screening of phage display libraries.
Human antibodies are also produced by immunization of a non-human transgenic animal containing some or all of the human immunoglobulin heavy and light chain loci, e.g., xenoMouse, in its genome with human IgE antigen TM Animals (Abgenix, inc./amben, inc. - -Fremont, calif.). XenoMouse TM Mice are engineered mouse lines that contain large fragments of human immunoglobulin heavy and light chain loci and are deficient in mouse antibody production. See, e.g., green et al, nature Genetics,7:13-21,1994; and U.S. Pat. nos. 5,916,771, 5,939,598, 5,985,615, 5,998,209, 6,075,181, 6,091,001, 6,114,598, 6,130,364, 6,162,963, and 6,150,584. See also WO 91/10741, WO 94/02602, WO 96/34096, WO 96/33735, WO 98/16654, WO 98/24893, WO 98/50433, WO 99/45031, WO 99/53049, WO 00/09560, and WO 00/037504.XenoMouse TM Mice produce adult-like human reservoirs of fully human antibodies (add-like human repertoire), and produce antigen-specific human antibodies. In some embodiments, xenoMouse is created by introducing megabase-sized, germline configuration fragments of the human heavy chain locus and the kappa light chain locus in Yeast Artificial Chromosomes (YACs) TM Mice contain approximately 80% of the human antibody V gene reservoir. In other embodiments, xenoMouse TM Mice also contain approximately all human lambda light chain loci. See Mendez et al, nature Genetics,15:146-156,1997; green and Jakobovits, J.Exp. Med.188:483-495 (1998) and WO 98/24893 (each incorporated by reference in its entirety for the purpose of teaching the preparation of fully human antibodies). In another aspect, the invention provides methods for preparing anti-PD-L1 antibodies from non-human, non-mouse animals by immunizing a non-human transgenic animal comprising a human immunoglobulin locus with a PD-L1 antigen. One can produce such animals using the methods described in the documents mentioned above.
Characterization of antibody binding to antigen
The binding of the antibodies of the invention to PD-L1 can be tested by, for example, standard ELISA. As one example, microtiter plates were coated with purified PD-L1 in PBS and then blocked with 5% bovine serum albumin in PBS. Dilutions of antibodies (e.g., dilutions of plasma from PD-L1 immunized mice) were added to each well and incubated at 37 ℃ for 1-2 hours. Plates were washed with PBS/Tween and then incubated with a second reagent conjugated with alkaline phosphatase (e.g., goat anti-human IgG Fc specific polyclonal reagent for human antibodies) at 37 ℃ for 1 hour. After washing, the plates were visualized with pNPP substrate (1 mg/ml) and analyzed at OD 405-650. Preferably, the mice producing the highest titers will be used for fusion. ELISA assays can also be used to screen hybridomas that show positive reactivity with PD-L1 immunogens. Hybridomas that bind to PD-L1 with high avidity are subcloned and further characterized. One clone that retains the reactivity of the parent cells (by ELISA) can be selected from each hybridoma for the preparation of 5-10 vial cell banks stored at-140 ℃ and for antibody purification.
To determine whether a selected anti-PD-L1 monoclonal antibody binds to a unique epitope, each antibody may be biotinylated using commercially available reagents (Pierce, rockford, ill.). Competition studies using unlabeled monoclonal antibodies and biotinylated monoclonal antibodies can be performed using PD-L1 coated-ELISA plates as described above. Biotinylated mAb binding can be detected using a streptavidin-alkaline phosphatase (strep-avidin-alkaline phosphatase) probe. To determine the isotype of a purified antibody, an isotype ELISA can be performed using reagents specific for antibodies of a particular isotype. For example, to determine the isotype of human monoclonal antibodies, wells of a microtiter plate may be coated with 1 μg/ml of anti-human immunoglobulin overnight at 4 ℃. After blocking with 1% bsa, the plates were reacted with 1 μg/ml or less of the test monoclonal antibody or purified isotype control at ambient temperature for 1 hour to 2 hours. The wells may then be reacted with a human IgG1 or human IgM specific alkaline phosphatase conjugated probe. The plates were developed and analyzed as described above.
anti-PD-L1 human IgG can be further tested for reactivity with PD-L1 antigen by Western blotting. Briefly, PD-L1 can be prepared and PD-L1 subjected to sodium dodecyl sulfate polyacrylamide gel electrophoresis. After electrophoresis, the isolated antigen was transferred to nitrocellulose membrane, blocked with 10% fetal bovine serum, and probed with the monoclonal antibody to be tested. Human IgG binding can be detected using anti-human IgG alkaline phosphatase and developed with BCIP/NBT substrate tablets (Sigma Chem.Co., st.Louis, mo.).
Identification of anti-PD-L1 antibodies
The present invention provides monoclonal antibodies and antigen binding fragments thereof that specifically bind to PD-L1 antigen.
Also included in the invention are antibodies that bind to the same epitope as the anti-PD-L1 antibodies of the invention. To determine whether an antibody can compete for binding to the same epitope as that bound by the anti-PD-L1 antibodies of the invention, a cross-blocking assay, such as a competitive ELISA assay, can be performed. In an exemplary competitive ELISA assay, PD-L1 coated on wells of a microtiter plate is pre-incubated with or without candidate competitive antibodies, and then the biotin-labeled anti-PD-L1 antibodies of the invention are added. The amount of labeled anti-PD-L1 antibody that binds to PD-L1 antigen in the well is measured using avidin-peroxidase conjugate and appropriate substrate. The antibody may be labeled with a radioactive or fluorescent label or some other detectable and measurable label. The amount of labeled anti-PD-L1 antibody that binds to an antigen will have an indirect correlation with the ability of the candidate competing antibody (test antibody) to compete for binding to the same epitope, i.e., the greater the affinity of the test antibody for the same epitope, the less the labeled antibody will bind to the antigen-coated well. A candidate competing antibody is considered to be an antibody that binds substantially to the same epitope or competes for binding to the same epitope as an anti-PD-L1 antibody of the invention if the candidate antibody can block binding of the PD-L1 antibody by at least 20%, preferably by at least 20-50%, even more preferably by at least 50% compared to a control run in parallel in the absence of the candidate competing antibody. It will be appreciated that variations of this assay can be performed to achieve the same quantitative value. In various embodiments, the antibodies of the invention include antibodies that bind to the same epitope as the monoclonal antibody designated pdl1#1. In various embodiments, the antibodies of the invention include antibodies that bind to the same epitope as the monoclonal antibody designated pdl1#10.
The amino acid sequences of the heavy and light chain CDRs of two murine antibodies pdl1#1 and pdl1#10 generated as described herein are shown in table 2 below.
TABLE 2
Heavy chain CDR
Light chain CDR
In various embodiments of the invention, the antibody or antigen binding fragment is a murine antibody pdl1#1 comprising the heavy chain variable region sequence of SEQ ID No. 12, and the light chain variable region sequence of SEQ ID No. 16, SEQ ID No. 12:
SEQ ID NO. 16 is:
in certain alternative embodiments, the antibody is an antibody comprising a heavy chain and a light chain, wherein the heavy chain comprises a heavy chain variable region, and wherein the heavy chain variable region comprises a sequence having at least about 80%, at least about 85%, at least about 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or at least about 99% identity to the amino acid sequence set forth in SEQ ID No. 12 or a polynucleotide sequence corresponding thereto SEQ ID No. 13, SEQ ID No. 13:
and wherein the light chain comprises a light chain variable region, and wherein the light chain variable region comprises a sequence having at least about 75%, at least about 80%, at least about 85%, at least about 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98% or at least about 99% identity to the amino acid sequence set forth in SEQ ID No. 16 or a polynucleotide sequence corresponding thereto SEQ ID No. 17, SEQ ID No. 17 being:
In various embodiments of the invention, the antibody or antigen binding fragment is a murine antibody pdl1#10 comprising the heavy chain variable region sequence of SEQ ID No. 14, and the light chain variable region sequence of SEQ ID No. 18, SEQ ID No. 14:
SEQ ID NO. 18 is:
in certain alternative embodiments, the antibody is an antibody comprising a heavy chain and a light chain, wherein the heavy chain comprises a heavy chain variable region, and wherein the heavy chain variable region comprises a sequence having at least about 80%, at least about 85%, at least about 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or at least about 99% identity to the amino acid sequence set forth in SEQ ID No. 14 or a polynucleotide sequence corresponding thereto SEQ ID No. 15, SEQ ID No. 15:
and wherein the light chain comprises a light chain variable region, and wherein the light chain variable region comprises a sequence having at least about 75%, at least about 80%, at least about 85%, at least about 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98% or at least about 99% identity to the amino acid sequence set forth in SEQ ID No. 18 or a polynucleotide sequence corresponding thereto SEQ ID No. 19:
in various embodiments, an isolated humanized antibody or antigen-binding fragment thereof of the invention binds to human PD-L1 and comprises a heavy chain variable region having a sequence that is the same, substantially the same or substantially similar to SEQ ID NOS 20, 22, 24, 26, 28, 30, 32, 34, 36, 38, 40, 42, 44 and 46 and a light chain variable region having a sequence that is the same, substantially the same or substantially similar to SEQ ID NOS 21, 23, 25, 27, 29, 31, 35, 37, 39, 41, 43, 45 and 47.
In various embodiments, an antibody or antigen binding fragment thereof comprises a heavy chain variable domain comprising a sequence that differs from the sequence of a heavy chain variable domain having the amino acid sequence set forth in SEQ ID NOs 20, 22, 24, 26, 28, 30, 32, 34, 36, 38, 40, 42, 44, or 46 by only 15, 14, 13, 12, 11, 10, 9, 8, 7, 6, 5, 4, 3, 2, 1, or 0 residues, wherein each such sequence difference is independently a deletion, insertion, or substitution of an amino acid residue. In various embodiments, an antibody or antigen binding fragment thereof comprises a heavy chain variable domain comprising a sequence having at least about 80%, at least about 85%, at least about 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98% or at least about 99% identity to an amino acid sequence as set forth in SEQ ID NOs 20, 22, 24, 26, 28, 30, 32, 34, 36, 38, 40, 42, 44 or 46.
In various embodiments, an isolated humanized antibody or antigen binding fragment thereof of the invention binds to human PD-L1 and comprises a sequence that differs from a sequence having the light chain variable domain set forth in SEQ ID No. 21, 23, 25, 27, 29, 31, 33, 35, 37, 39, 41, 43, 45, or 47 by only 15, 14, 13, 12, 11, 10, 9, 8, 7, 6, 5, 4, 3, 2, 1, or 0 residues, wherein each such sequence difference is independently a deletion, insertion, or substitution of an amino acid residue. In various embodiments, an antibody or antigen binding fragment thereof comprises a light chain variable domain comprising a sequence having at least about 80%, at least about 85%, at least about 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98% or at least about 99% identity to an amino acid sequence as set forth in SEQ ID NOs 21, 23, 25, 27, 29, 31, 33, 35, 37, 39, 41, 43, 45 or 47.
In another embodiment, the heavy chain variable domain comprises a sequence of amino acids encoded by a polynucleotide that hybridizes under moderately stringent conditions to the complement (complement) of a polynucleotide encoding a heavy chain variable domain having the sequence of SEQ ID NO. 13 or 15. In another embodiment, the heavy chain variable domain comprises a sequence of amino acids encoded by a polynucleotide that hybridizes under stringent conditions to the complement of a polynucleotide encoding a heavy chain variable domain having the sequence of SEQ ID NO. 13 or 15. In another embodiment, the light chain variable domain comprises an amino acid sequence encoded by a polynucleotide that hybridizes under moderately stringent conditions to the complement of a polynucleotide encoding a light chain variable domain having the sequence of SEQ ID NO. 17 or 19. In another embodiment, the light chain variable domain comprises an amino acid sequence encoded by a polynucleotide that hybridizes under stringent conditions to the complement of a polynucleotide encoding a light chain variable domain having the sequence of SEQ ID NO. 17 or 19.
In various embodiments, the antibody is a humanized antibody comprising a heavy chain variable region having the amino acid sequence set forth in SEQ ID NO. 20 and a light chain variable region having the amino acid sequence set forth in SEQ ID NO. 21. In various embodiments, the antibody is a humanized antibody comprising a heavy chain variable region having the amino acid sequence set forth in SEQ ID NO. 22 and a light chain variable region having the amino acid sequence set forth in SEQ ID NO. 23. In various embodiments, the antibody is a humanized antibody comprising a heavy chain variable region having the amino acid sequence set forth in SEQ ID NO. 24 and a light chain variable region having the amino acid sequence set forth in SEQ ID NO. 25. In various embodiments, the antibody is a humanized antibody comprising a heavy chain variable region having the amino acid sequence set forth in SEQ ID NO. 26 and a light chain variable region having the amino acid sequence set forth in SEQ ID NO. 27. In various embodiments, the antibody is a humanized antibody comprising a heavy chain variable region having the amino acid sequence set forth in SEQ ID NO. 28 and a light chain variable region having the amino acid sequence set forth in SEQ ID NO. 29.
In various embodiments of the present disclosure, the antibody may be an anti-PD-L1 antibody having an antigen-binding affinity that is the same as or higher than an antigen-binding affinity of an antibody comprising a heavy chain sequence as set forth in any one of SEQ ID NOs 55, 57, 59, 61, and 63. In various embodiments, the antibody may be an anti-PD-L1 antibody that binds to the same epitope as an antibody comprising a heavy chain sequence as set forth in any one of SEQ ID NOs 55, 57, 59, 61 and 63. In various embodiments, the antibody is an anti-PD-L1 antibody that competes with an antibody comprising a heavy chain sequence as set forth in any one of SEQ ID NOs 55, 57, 59, 61 and 63. In various embodiments, the antibody may be an anti-PD-L1 antibody comprising at least one (such as 2 or 3) CDRs of a heavy chain sequence as set forth in any one of SEQ ID NOs 55, 57, 59, 61 and 63.
In various embodiments, the antibody comprises an amino acid sequence that shares, for example, at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, or at least 99% observed homology with any of SEQ ID NOs 55, 57, 59, 61, and 63. In various embodiments, an antibody comprises a nucleic acid sequence sharing, for example, at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, or at least 99% observed homology with any of SEQ ID NOs 65, 67, 69, 71, and 73.
In various embodiments of the present disclosure, the antibody may be an anti-PD-L1 antibody having an antigen-binding affinity that is the same as or higher than an antigen-binding affinity of an antibody comprising a light chain sequence as set forth in any one of SEQ ID NOs 56, 58, 60, 62, and 64. In various embodiments, the antibody may be an anti-PD-L1 antibody that binds to the same epitope as an antibody comprising a light chain sequence as set forth in any one of SEQ ID NOs 56, 58, 60, 62 and 64. In various embodiments, the antibody is an anti-PD-L1 antibody that competes with an antibody comprising a light chain sequence as set forth in any one of SEQ ID NOs 56, 58, 60, 62 and 64. In various embodiments, the antibody may be an anti-PD-L1 antibody comprising at least one (such as 2 or 3) CDRs of the light chain sequences as set forth in any one of SEQ ID NOs 56, 58, 60, 62 and 64.
In various embodiments, the antibody comprises an amino acid sequence that shares, for example, at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, or at least 99% observed homology with any of SEQ ID NOs 56, 58, 60, 62, and 64. In various embodiments, an antibody comprises a nucleic acid sequence that shares, for example, at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, or at least 99% observed homology with any of SEQ ID NOs 66, 68, 70, 72, and 74.
In various embodiments, an isolated humanized antibody or antigen binding fragment thereof of the invention binds to human PD-L1 and comprises the heavy chain sequence set forth in SEQ ID NO. 55 and the light chain sequence set forth in SEQ ID NO. 56.
In various embodiments, an isolated humanized antibody or antigen binding fragment thereof of the invention binds to human PD-L1 and comprises the heavy chain sequence set forth in SEQ ID NO:57 and the light chain sequence set forth in SEQ ID NO: 58.
In various embodiments, an isolated humanized antibody or antigen binding fragment thereof of the invention binds to human PD-L1 and comprises the heavy chain sequence set forth in SEQ ID NO. 59 and the light chain sequence set forth in SEQ ID NO. 60.
In various embodiments, an isolated humanized antibody or antigen binding fragment thereof of the invention binds to human PD-L1 and comprises the heavy chain sequence set forth in SEQ ID NO. 61 and the light chain sequence set forth in SEQ ID NO. 62.
In various embodiments, an isolated humanized antibody or antigen binding fragment thereof of the invention binds to human PD-L1 and comprises the heavy chain sequence set forth in SEQ ID NO. 63 and the light chain sequence set forth in SEQ ID NO. 64.
The antibodies or antigen binding fragments thereof of the invention may comprise any constant region known in the art. The light chain constant region may be, for example, a kappa or lambda type light chain constant region, such as a human kappa or lambda type light chain constant region. The heavy chain constant region may be, for example, an alpha, delta, epsilon, gamma or mu type heavy chain constant region, such as IgA, igD, igE, igG and IgM type heavy chain constant regions. In various embodiments, the light chain constant region or heavy chain constant region is a fragment, derivative, variant, or mutein (mutein) of a naturally occurring constant region.
Techniques for deriving antibodies of different subclasses or isotypes from antibodies of interest, i.e., subclass switching (subclass switching), are known. Thus, igG antibodies may be derived from, for example, igM antibodies, and vice versa. Such techniques allow the preparation of new antibodies having the antigen binding properties of a given antibody (parent antibody) but also exhibiting biological properties associated with an antibody isotype or subclass that differs from the parent antibody. Recombinant DNA techniques may be used. Cloned DNA encoding a particular antibody polypeptide, e.g., DNA encoding the constant domains of antibodies of the desired isotype, may be used in such procedures. See also Lanitto et al Methods mol. Biol.178:303-16,2002.
In various embodiments, the antibodies of the invention further comprise a light chain kappa or lambda constant domain or fragment thereof, and further comprise a heavy chain constant domain or fragment thereof. The sequences of the light chain constant region and the heavy chain constant region used in the exemplified antibodies and polynucleotides encoding them are provided below.
Light chain (kappa) constant region
Light chain (lambda) constant region
Heavy chain constant region
Antibodies of the invention may also be described or designated in terms of their cross-reactivity. Antibodies that bind to PD-L1 polypeptides that have at least 95%, at least 90%, at least 85%, at least 80%, at least 75%, at least 70%, at least 65%, at least 60%, at least 55%, and at least 50% identity (as calculated using methods known in the art and described herein) to human PD-L1 are also included in the invention.
The invention also includes antibodies that bind to the same epitope as the anti-PD-L1 antibodies of the invention. To determine whether an antibody can compete for binding to the same epitope as that bound by the anti-PD-L1 antibodies of the invention, a cross-blocking assay, such as a competitive ELISA assay, can be performed. In an exemplary competitive ELISA assay, PD-L1 coated on wells of a microtiter plate is pre-incubated with or without candidate competitive antibodies, and then the biotin-labeled anti-PD-L1 antibodies of the invention are added. The amount of labeled anti-PD-L1 antibody that binds to PD-L1 antigen in the well is measured using avidin-peroxidase conjugate and appropriate substrate. The antibody may be labeled with a radioactive or fluorescent label or some other detectable and measurable label. The amount of labeled anti-PD-L1 antibody that binds to an antigen will have an indirect correlation with the ability of the candidate competing antibody (test antibody) to compete for binding to the same epitope, i.e., the greater the affinity of the test antibody for the same epitope, the less the labeled antibody will bind to the antigen-coated well. A candidate competing antibody is considered to be an antibody that binds substantially to the same epitope or competes for binding to the same epitope as an anti-PD-L1 antibody of the invention if the candidate antibody can block binding of the PD-L1 antibody by at least 20%, at least 30%, at least 40% or at least 50% compared to a control run in parallel in the absence of the candidate competing antibody. It will be appreciated that variations of this assay can be performed to achieve the same quantitative value.
In various embodiments of the invention, the antibody or antigen-binding fragment is a murine-human chimeric antibody derived from murine pdl1#10, said antibody or antigen-binding fragment comprising the heavy chain sequence of SEQ ID No. 51 and the light chain sequence of SEQ ID No. 53, and wherein amino acids 1-19 of SEQ ID No. 51 are the leader sequences:
and wherein amino acids 1-19 of SEQ ID NO. 53 are the leader sequence:
in certain alternative embodiments, the antibody is a murine-human chimeric antibody comprising a heavy chain and a light chain, wherein the heavy chain comprises a sequence having at least about 80%, at least about 85%, at least about 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or at least about 99% identity to the amino acid sequence set forth in SEQ ID No. 51 or its corresponding polynucleotide sequence SEQ ID No. 52:
and wherein the light chain comprises a sequence having at least about 75%, at least about 80%, at least about 85%, at least about 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or at least about 99% identity to the amino acid sequence set forth in SEQ ID No. 53 or a polynucleotide sequence corresponding thereto SEQ ID No. 54:
in various embodiments of the invention, the antibody is a humanized IgG comprising the heavy chain sequence of SEQ ID No. 55 and the light chain sequence of SEQ ID No. 56, and wherein amino acids 1-19 of SEQ ID No. 55 are the leader sequence:
And wherein amino acids 1-19 of SEQ ID NO. 56 are the leader sequence:
in certain alternative embodiments, the antibody is an antibody comprising a heavy chain and a light chain, wherein the heavy chain comprises a sequence having at least about 80%, at least about 85%, at least about 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98% or at least about 99% identity to the amino acid sequence set forth in SEQ ID No. 55 or a polynucleotide sequence corresponding thereto SEQ ID No. 65, SEQ ID No. 65:
and wherein the light chain comprises a sequence having at least about 75%, at least about 80%, at least about 85%, at least about 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or at least about 99% identity to the amino acid sequence set forth in SEQ ID No. 56 or a polynucleotide sequence corresponding thereto, SEQ ID No. 66 is:
in various embodiments of the invention, the antibody is a humanized IgG comprising the heavy chain sequence of SEQ ID NO. 57 and the light chain sequence of SEQ ID NO. 58, and wherein amino acids 1-19 of SEQ ID NO. 57 are the leader sequence:
and wherein amino acids 1-19 of SEQ ID NO. 58 are the leader sequence:
in certain alternative embodiments, the antibody is an antibody comprising a heavy chain and a light chain, wherein the heavy chain comprises a sequence having at least about 80%, at least about 85%, at least about 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98% or at least about 99% identity to the amino acid sequence set forth in SEQ ID No. 57 or a polynucleotide sequence corresponding thereto SEQ ID No. 67, SEQ ID No. 67:
And wherein the light chain comprises a sequence having at least about 75%, at least about 80%, at least about 85%, at least about 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or at least about 99% identity to the amino acid sequence set forth in SEQ ID No. 58 or a polynucleotide sequence corresponding thereto SEQ ID No. 68:
in various embodiments of the invention, the antibody is a humanized IgG comprising the heavy chain sequence of SEQ ID No. 59 and the light chain sequence of SEQ ID No. 60, and wherein amino acids 1-19 of SEQ ID No. 59 are the leader sequence:
and wherein amino acids 1-19 of SEQ ID NO. 60 are the leader sequence:
in certain alternative embodiments, the antibody is an antibody comprising a heavy chain and a light chain, wherein the heavy chain comprises a sequence having at least about 80%, at least about 85%, at least about 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98% or at least about 99% identity to the amino acid sequence set forth in SEQ ID No. 59 or a polynucleotide sequence corresponding thereto SEQ ID No. 69, SEQ ID No. 69 being:
and wherein the light chain comprises a sequence having at least about 75%, at least about 80%, at least about 85%, at least about 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or at least about 99% identity to the amino acid sequence set forth in SEQ ID No. 60 or a polynucleotide sequence corresponding thereto SEQ ID No. 70:
In various embodiments of the invention, the antibody is a humanized IgG comprising the heavy chain sequence of SEQ ID No. 61 and the light chain sequence of SEQ ID No. 62, and wherein amino acids 1-19 of SEQ ID No. 61 are the leader sequence:
and wherein amino acids 1-19 of SEQ ID NO. 62 are the leader sequence:
/>
in certain alternative embodiments, the antibody is an antibody comprising a heavy chain and a light chain, wherein the heavy chain comprises a sequence having at least about 80%, at least about 85%, at least about 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98% or at least about 99% identity to the amino acid sequence set forth in SEQ ID No. 61 or a polynucleotide sequence corresponding thereto SEQ ID No. 71, SEQ ID No. 71:
and wherein the light chain comprises a sequence having at least about 75%, at least about 80%, at least about 85%, at least about 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or at least about 99% identity to the amino acid sequence as set forth in SEQ ID No. 62 or a polynucleotide sequence corresponding thereto SEQ ID No. 72:
in various embodiments of the invention, the antibody is a humanized IgG comprising the heavy chain sequence of SEQ ID No. 63 and the light chain sequence of SEQ ID No. 64, and wherein amino acids 1-19 of SEQ ID No. 63 are the leader sequence:
And wherein amino acids 1-19 of SEQ ID NO. 64 are the leader sequence:
in certain alternative embodiments, the antibody is an antibody comprising a heavy chain and a light chain, wherein the heavy chain comprises a sequence having at least about 80%, at least about 85%, at least about 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98% or at least about 99% identity to the amino acid sequence set forth in SEQ ID No. 63 or a polynucleotide sequence corresponding thereto SEQ ID No. 73, SEQ ID No. 73 being:
and wherein the light chain comprises a sequence having at least about 75%, at least about 80%, at least about 85%, at least about 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or at least about 99% identity to the amino acid sequence set forth in SEQ ID No. 64 or a polynucleotide sequence corresponding thereto SEQ ID No. 74:
in certain alternative embodiments, antibodies of the invention may be produced by modifying one or both of the variable regions (i.e., V H And/or V L ) One or more residues in (a), or by modification of one or moreResidues in multiple constant regions are engineered, for example, to alter one or more effector functions of an antibody. In various embodiments, the variable regions of the antibodies will be modified by CDR grafting using framework sequences that can be obtained from public DNA databases including germline antibody gene sequences or published references (e.g., tomlinson, I.M., et al, J. Mol. Biol.227:776-798,1992; and Cox, J. P.L. Et al, eur. J. Immunol.24:827-836,1994; the contents of each of which are expressly incorporated herein by reference). In various embodiments, antibodies may be modified using site-directed mutagenesis or PCR-mediated mutagenesis to introduce one or more mutations in VH and/or VL that improve binding affinity and/or reduce immunogenicity. In various embodiments, antibodies can be modified in the Fc region for the purpose of altering the serum half-life of the antibody, complement fixation, fc receptor binding, and/or antigen-dependent cytotoxicity. In various embodiments, the antibodies may be modified for the purpose of modifying glycosylation of the antibodies. Methods for performing each modification described herein, as well as other methods, are well known to those skilled in the art.
Pharmaceutical composition
In another aspect, the invention provides a pharmaceutical composition comprising an antibody or antigen-binding fragment thereof as described above. The pharmaceutical compositions, methods and uses of the present invention thus also include embodiments in combination (co-administration) with other active agents, as detailed below.
Generally, the antibodies or antigen-binding fragments thereof of the invention are suitable for administration as a formulation in combination with one or more pharmaceutically acceptable excipients. The term "excipient" is used herein to describe any ingredient other than one or more compounds of the present invention. The choice of one or more excipients will depend largely on factors such as the particular mode of administration, the effect of the excipient on solubility and stability, and the nature of the dosage form. As used herein, "pharmaceutically acceptable excipients" include any and all solvents, dispersion media, coatings, antibacterial and antifungal agents, isotonic and absorption delaying agents, and the like that are physiologically compatible. Some examples of pharmaceutically acceptable excipients are water, saline, phosphate buffered saline, dextrose, glycerol, ethanol, and the like, and combinations thereof. In many cases, it will be preferable to include isotonic agents, for example, sugars, polyalcohols such as mannitol, sorbitol, or sodium chloride in the composition. Further examples of pharmaceutically acceptable substances are wetting agents or minor amounts of auxiliary substances such as wetting or emulsifying agents, preservatives or buffers which prolong the shelf life of the antibody or enhance the efficacy of the antibody. The pharmaceutical compositions of the present invention and methods for their preparation will be readily apparent to those skilled in the art. Such compositions and methods for their preparation can be found, for example, in Remington, pharmaceutical Sciences, 19 th edition (Mack Publishing Company, 1995). The pharmaceutical composition is preferably manufactured under GMP conditions.
The pharmaceutical compositions of the present invention may be prepared, packaged or marketed in bulk in single unit doses or in more than one single unit dose. As used herein, a "unit dose" is a discrete amount of a pharmaceutical composition comprising a predetermined amount of an active ingredient. The amount of active ingredient is typically equal to the dose of active ingredient to be administered to the subject or a convenient fraction of such dose, for example half or one third of such dose.
Any method recognized in the art for administering a peptide, protein, or antibody may be suitably employed with the antibodies and portions of the invention.
The pharmaceutical compositions of the present invention are generally suitable for parenteral administration. As used herein, "parenteral administration" of a pharmaceutical composition includes any route of administration characterized by: the tissue of the subject is physically perforated (physical breaching) and the pharmaceutical composition is administered through a gap (break) in the tissue, thus generally causing direct administration into the blood stream, into muscles or into internal organs. Thus, parenteral administration includes, but is not limited to, administration of pharmaceutical compositions by injection of the composition, application of the composition by surgical incision, application of the composition by tissue-permeable non-surgical wound, and the like. In particular, parenteral administration is contemplated to include, but are not limited to, subcutaneous, intraperitoneal, intramuscular, intrasternal, intravenous, intraarterial, intrathecal, intraventricular, intraurethral, intracranial, intrasynovial injection or infusion; and kidney dialysis infusion techniques. Various embodiments include intravenous and subcutaneous routes.
Formulations of pharmaceutical compositions suitable for parenteral administration often comprise the active ingredient in combination with a pharmaceutically acceptable carrier such as sterile water or sterile isotonic saline. Such formulations may be prepared, packaged or sold in a form suitable for bolus administration (bolus administration) or for continuous administration. The injectable formulations may be prepared, packaged or sold in unit dosage forms, such as in ampoules or in multi-dose containers containing a preservative. Formulations for parenteral administration include, but are not limited to, suspensions, solutions, emulsions in oily or aqueous vehicles, pastes, and the like. Such formulations may also contain one or more additional ingredients including, but not limited to, suspending, stabilizing or dispersing agents. In one embodiment of a formulation for parenteral administration, the active ingredient is provided in dry (i.e., powder or granule) form for reconstitution with a suitable vehicle (e.g., sterile pyrogen-free water) and then the reconstituted composition is administered parenterally. Parenteral formulations also include aqueous solutions which may contain excipients such as salts, carbohydrates and buffers (preferably to a pH of from 3 to 9), but for some applications they may be more suitable to be formulated as sterile non-aqueous solutions or in dry form for use with a suitable vehicle such as sterile, pyrogen-free water. Exemplary forms of parenteral administration include solutions or suspensions in sterile aqueous solutions, such as aqueous propylene glycol or dextrose solutions. Such dosage forms may be suitably buffered if desired. Other useful parenterally administrable formulations include those comprising the active ingredient in microcrystalline form, or in liposome preparations. Formulations for parenteral administration may be formulated for immediate release and/or modified release (modified release). Modified release formulations include delayed, sustained, pulsed, controlled, targeted and programmed release.
For example, in one aspect, a sterile injectable solution can be prepared by incorporating the anti-PD-L1 antibody in the required amount in an appropriate solvent with one or a combination of ingredients enumerated above, as required, followed by filtered sterilization. Generally, the dispersing agents are prepared by incorporating the active compound into a sterile vehicle which contains a basic dispersion medium and the required other ingredients from those enumerated above. In the case of sterile powders for the preparation of sterile injectable solutions, the preferred methods of preparation are vacuum drying and freeze-drying which yield a powder of the active ingredient plus any additional desired ingredient from a previously sterile-filtered solution thereof. Proper solution fluidity can be maintained, for example, by the use of a coating such as lecithin, by the maintenance of the required particle size in the case of dispersions and by the use of surfactants. Prolonged absorption of the injectable compositions can be brought about by including in the composition agents which delay absorption, for example, monostearates and gelatins.
The antibodies of the invention may also be administered intranasally or by inhalation, typically in the form of a dry powder (alone, as a mixture or as mixed component particles, e.g. mixed with a suitable pharmaceutically acceptable excipient) from a dry powder inhaler, as an aerosol spray from a pressure vessel, pump, nebulizer (spray), nebulizer (preferably a nebulizer using electrohydrodynamic to produce a fine mist) or nebulizer (nebulizer), with or without a suitable propellant, or as nasal drops.
Pressure vessels, pumps, nebulizers, or nebulizers (nebulizers) typically comprise solutions or suspensions of the antibodies of the invention containing, for example, a suitable agent for dispersing, dissolving or prolonging the release of the active agent, one or more propellants as solvents.
Prior to use in dry powder or suspension formulations, the pharmaceutical product is typically micronized to a size suitable for delivery by inhalation (typically less than 5 microns). This may be done by any suitable comminution method, such as spiral jet milling, fluid bed jet milling, supercritical fluid processing to form nanoparticles, high pressure homogenization or spray drying.
Capsules, blisters and cartridges for use in an inhaler or insufflator (insuffator) may be formulated containing a powder mix of a compound of the invention, a suitable powder base and a performance modifier (performance modifier).
Suitable flavouring agents (flavour) such as menthol and left menthol, or sweetening agents such as saccharin or sodium saccharin, may be added to those formulations of the invention intended for inhalation/intranasal administration.
Formulations for inhalation/intranasal administration may be formulated for immediate release and/or modified release. Modified release formulations include delayed, sustained, pulsed, controlled, targeted and programmed release.
In the case of dry powder inhalers and aerosols, the dosage unit is determined by means of a valve delivering a metered amount. The units according to the invention are typically arranged to administer a metered dose or "puff" of the antibodies of the invention. The total daily dose will typically be administered as a single dose, or more typically, as divided doses throughout the day.
The antibodies and antibody portions of the invention may also be formulated for administration by the oral route. Oral administration may involve swallowing, such that the compound enters the gastrointestinal tract, and/or buccal, lingual or sublingual administration, by which means the compound enters the blood stream directly from the mouth.
Formulations suitable for oral administration include solid, semi-solid, and liquid systems, such as tablets; soft or hard capsules comprising multiparticulates or nanoparticles, liquids or powders; lozenges (including liquid-filled); chewing agents (chews); gel; a fast-dispersing dosage form; a film; oval agent (ovile); sprays and buccal/mucoadhesive patches (buccal/mucoadhesive patch).
Pharmaceutical compositions intended for oral use may be prepared according to any method known in the art for manufacturing pharmaceutical compositions, and such compositions may comprise one or more agents selected from the group consisting of sweeteners to provide pharmaceutically elegant (elegant) and palatable preparations. For example, to prepare an orally deliverable tablet, the antibody or antigen-binding fragment thereof is admixed with at least one pharmaceutical excipient and the solid formulation is compressed according to known methods to form a tablet for delivery to the gastrointestinal tract. Tablet compositions are typically formulated with additives such as saccharide or cellulosic carriers, binders such as starch paste or methylcellulose, fillers, disintegrants or other additives commonly used in the manufacture of medical formulations. To prepare an orally deliverable capsule, DHEA is mixed with at least one pharmaceutical excipient and the solid formulation is placed in a capsule container suitable for delivery to the gastrointestinal tract. Compositions comprising antibodies or antigen binding fragments thereof may be prepared as generally described in Remington, pharmaceutical Sciences, 18 th edition 1990 (Mack Publishing co.easton pa.18042), incorporated herein by reference.
In various embodiments, the pharmaceutical composition is formulated as an orally deliverable tablet comprising the antibody or antigen-binding fragment thereof admixed with a non-toxic pharmaceutically acceptable excipient suitable for use in the manufacture of a tablet. These excipients may be inert diluents such as calcium carbonate, sodium carbonate, lactose, calcium phosphate or sodium phosphate; granulating and disintegrating agents, for example, corn starch, gelatin or acacia, and lubricating agents, for example, magnesium stearate, stearic acid or talc. The tablets may be uncoated or they may be coated by known techniques to delay disintegration and absorption in the gastrointestinal tract and thereby provide a sustained action over a longer period. For example, a time delay material such as glyceryl monostearate or glyceryl distearate alone or with a wax may be employed.
In various embodiments, the pharmaceutical composition is formulated as a hard gelatin capsule wherein the antibody or antigen-binding fragment thereof is mixed with an inert solid diluent, such as calcium carbonate, calcium phosphate or kaolin, or as a soft gelatin capsule wherein the antibody or antigen-binding fragment thereof is mixed with an aqueous or oil medium, such as peanut oil (arachis oil), peanut oil (peanut oil), liquid paraffin or olive oil.
Liquid preparations include suspensions, solutions, syrups and elixirs. Such formulations may be used as fillers in soft or hard capsules (made from, for example, gelatin or hydroxypropyl methylcellulose) and typically comprise a carrier such as water, ethanol, polyethylene glycol, propylene glycol, methylcellulose or a suitable oil, together with one or more emulsifying and/or suspending agents. Liquid formulations may also be prepared by reconstitution of solids, for example from a sachet.
Therapeutic use
In another aspect, the invention relates to a method for treating a subject suffering from a T cell-related disorder or a disorder that can be alleviated or prevented by boosting or suppressing or prolonging the immune response, the method comprising administering to the subject a therapeutically effective amount (as monotherapy or in a combination therapy regimen) of an antibody of the invention or an antigen-binding fragment thereof. In various embodiments, the subject is a human subject.
In another aspect, the invention provides a method for treating cancer in a subject, the method comprising administering to the subject a therapeutically effective amount (as monotherapy or in a combination therapy regimen) of an antibody of the invention or an antigen binding fragment thereof. Such disorders include, but are not limited to, solid tumors, bone cancer, pancreatic cancer, skin cancer, head and neck cancer (cancer of the head or neck), cutaneous or intraocular malignant melanoma, uterine cancer, ovarian cancer, rectal cancer, cancer of the anal region, gastric cancer, testicular cancer, uterine cancer, fallopian tube cancer (carcinoma of the fallopian tube), endometrial cancer, cervical cancer, vaginal cancer, vulvar cancer, hodgkin's Disease, non-Hodgkin's lymphoma, esophageal cancer, small intestine cancer, cancer of the endocrine system, thyroid cancer, parathyroid cancer, adrenal cancer, soft tissue sarcoma, urinary tract cancer, penile cancer, chronic or acute leukemia including acute myelogenous leukemia, chronic myelogenous leukemia, acute lymphocytic leukemia, chronic lymphocytic leukemia, childhood solid tumors, lymphocytic lymphomas, bladder cancer, renal cancer or ureteral cancer, renal pelvis cancer, neoplasms of the Central Nervous System (CNS), primary CNS lymphomas, tumor angiogenesis, spinal axis tumors (spinal axis tumor), brain stem glioma, pituitary adenoma, kaposi's sarcoma, epidermoid carcinoma, squamous cell carcinoma, T cell carcinoma, squamous cell carcinoma, and combinations thereof. In various embodiments, the subject previously responded to treatment with an anti-cancer therapy, but suffered from a relapse (hereinafter referred to as "recurrent cancer") after cessation of the therapy. In various embodiments, the subject has a resistant cancer or refractory cancer. In various embodiments, the cancer cells are immunogenic tumors (e.g., those tumors that may be vaccinated against tumor challenge using the tumor itself). In various embodiments, the cancer is selected from the group consisting of: melanoma (e.g., metastatic malignant melanoma), colorectal cancer (CRC), renal cancer, bladder cancer, non-small cell lung cancer (NSCLC), prostate cancer, breast cancer, colon cancer, ovarian cancer, and lung cancer.
In various embodiments, the antibodies of the invention, and antigen-binding fragments thereof, can be used to directly kill or ablate cancer cells in vivo. Direct killing involves administering an antibody (which is optionally fused with a cytotoxic drug) to a subject in need of such treatment. In various embodiments, the cancer comprises cancer cells that express PD-L1 at a higher level than non-cancerous cells of comparable tissue. Because antibodies recognize PD-L1 on cancer cells, any such cells that bind to the antibody are destroyed. In the case where antibodies are used alone to kill or ablate cancer cells, such killing or ablation may act by eliciting endogenous host immune functions such as CDC and/or ADCC. Assays for determining whether an antibody kills cells in this manner are within the purview of those skilled in the art.
In various embodiments, the antibodies and antigen binding fragments thereof of the invention may be used to promote inhibition of growth and/or proliferation of cancerous tumor cells. These methods can inhibit or prevent the growth of cancer cells in the subject, such as, for example, at least 10%, at least 15%, at least 20%, at least 25%, at least 30%, at least 35%, at least 40%, at least 45%, at least 50%, at least 55%, at least 60%, at least 65%, at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, or at least 95%. Thus, where the cancer is a solid tumor, the antibodies and antigen-binding fragments thereof of the invention can reduce the size of the solid tumor by at least 10%, at least 15%, at least 20%, at least 25%, at least 30%, at least 35%, at least 40%, at least 45%, at least 50%, at least 55%, at least 60%, at least 65%, at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, or at least 95%.
Inhibition of cancer cell proliferation may be measured by cell-based assays such as Bromodeoxyuridine (BRDU) incorporation (Hoshino et al, int. J. Cancer 38,369,1986; campana et al, J. Immunol. Meth.107:79,1988); [ 3 H]Thymidine incorporation (Chen, J., oncogene 13:1395-403,1996; jelong, J.; J.biol. Chem.270:18367-73,1995; dye Alamar Blue (available from Biosource International) (Voytik-Harbin et al In Vitro Cell Dev Biol Anim 34:239-46,1998). Anchorage-independent growth of cancer cells (anchorage independent growth) was assessed by colony formation assays in soft agar, such as by counting the number of cancer cell colonies formed on soft agar (see examples and Sambrook et al Molecular Cloning, cold Spring Harbor, 1989).
Inhibition of cancer cell growth in a subject can be assessed by monitoring cancer growth in the subject, e.g., in an animal model or in a human subject. One exemplary monitoring method is tumorigenicity determination. In one example, the xenograft comprises human cells from a pre-existing tumor or from a tumor cell line. Tumor xenograft assays are known in the art and are described herein (see, e.g., ogawa et al, oncogene 19:6043-6052,2000). In another embodiment, tumorigenic capacity is monitored using a hollow fiber assay, described in U.S. patent No. 5,698,413, which is incorporated herein by reference in its entirety.
The percentage of inhibition was determined by comparing the proliferation, anchoring independent growth or cancer of cancer cells under modulator treatment (modulator treatment) and under negative control conditions (usually without modulator treatment)Cell growth was calculated. For example, in the number of cancer cells or cancer cell colonies (colony formation assay), or PRDU or [ 3 H]In the case of thymidine incorporation as se:Sub>A (under treatment with modulator) and C (under negative control conditions), the percentage inhibition will be (C-se:Sub>A)/c×100%.
Examples of tumor cell lines derived from human tumors and useful in vitro and in vivo studies include, but are not limited to, leukemia cell lines (e.g., CCRF-CEM, HL-60 (TB), K-562, MOLT-4, RPM1-8226, SR, P388, and P388/ADR); non-small cell lung cancer cell lines (e.g., A549/ATCC, EKVX, HOP-62, HOP-92, NCI-H226, NCI-H23, NCI-H322M, NCI-H460, NCI-H522, and LXFL 529); small cell lung cancer cell lines (e.g., DMS 114 and SHP-77); colon cancer cell lines (e.g., COLO 205, HCC-2998, HCT-116, HCT-15, HT29, KM12, SW-620, DLD-1, and KM20L 2); central Nervous System (CNS) cancer cell lines (e.g., SF-268, SF-295, SF-539, SNB-19, SNB-75, U251, SNB-78, and XF 498); melanoma cell lines (e.g., LOX I MVI, MALME-3M, M, SK-MEL-2, SK-MEL-28, SK-MEL-5, UACC-257, UACC-62, RPMI-7951, and M19-MEL); ovarian cancer cell lines (e.g., IGROV1, OVCAR-3, OVCAR-4, OVCAR-5, OVCAR-8, and SK-OV-3); renal cancer cell lines (e.g., 786-0, A498, ACHN, CAKI-1, RXF 393, SN12C, TK-10, UO-31, RXF-631, and SN12K 1); prostate cancer cell lines (e.g., PC-3 and DU-145); breast cancer cell lines (e.g., MCF7, NCI/ADR-RES, MDA-MB-231/ATCC, HS 578T, MDA-MB-435, BT-549, T-47D, and MDA-MB-468); and thyroid cancer cell lines (e.g., SK-N-SH).
In another aspect, the invention provides a method for treating an infectious disease in a subject, the method comprising administering to the subject a therapeutically effective amount (as monotherapy or in a combination therapy regimen) of an isolated antibody or antigen binding fragment of the invention. In various embodiments, the subject has an infectious disease caused by a pathogenic virus. In various embodiments, the subject has an infectious disease caused by pathogenic bacteria. In various embodiments, the subject has an infectious disease caused by a pathogenic fungus. In various embodiments, the subject has an infectious disease caused by a pathogenic parasite. In various embodiments, the subject has an infectious disease that is resistant to treatment with a conventional vaccine or that is not effectively treated by treatment with a conventional vaccine.
"therapeutically effective amount" or "therapeutically effective dose" refers to the amount of therapeutic agent administered that will alleviate one or more symptoms of the disorder being treated to some extent.
The therapeutically effective dose can be determined by determining IC 50 Initial evaluation was from cell culture assays. The dose may then be formulated to achieve a combination of IC's in animal models, including as determined in cell culture 50 Is a circulating plasma concentration range of (c). Such information can be used to more accurately determine the dosage useful in humans. The level in plasma may be measured, for example, by HPLC. The exact composition, route of administration and dosage may be selected by a separate physician in view of the subject's condition.
The dosage regimen can be adjusted to provide the best desired response (e.g., a therapeutic response or a prophylactic response). For example, a single bolus may be administered, several divided doses (multiple or repeated or sustained) may be administered over time and the doses may be proportionally reduced or increased as indicated by the urgent need for the treatment context. To facilitate administration and dose consistency, it is particularly beneficial to formulate parenteral compositions in dosage unit form. Dosage unit form as used herein refers to physically discrete units suitable as unitary dosages for the mammalian subject to be treated; each unit contains a predetermined amount of active compound calculated to produce the desired therapeutic effect together with the required pharmaceutical carrier. The specifications of the dosage unit forms of the present disclosure will be largely determined by the unique characteristics of the antibody and the particular therapeutic or prophylactic effect to be achieved.
Thus, the skilled artisan will appreciate, based on the disclosure provided herein, that dosages and dosing regimens are adjusted according to methods well known in the therapeutic arts. That is, the maximum tolerable dose can be readily determined, and an effective amount to provide a detectable therapeutic benefit to the subject can also be determined, as can the time requirements for administration of each dose to provide a detectable therapeutic benefit to the subject. Thus, although certain dosages and administration regimens are exemplified herein, these examples are in no way limiting as to the dosages and administration regimens that may be provided to a subject in practicing the disclosure.
It should be noted that the dose value may vary with the type and severity of the condition to be alleviated, and may include a single dose or more than one dose. It will also be appreciated that for any particular subject, the particular dosage regimen should be adjusted over time according to the individual needs and the professional judgment of the person administering or supervising the administration of the compositions, and that the dosage ranges set forth herein are exemplary only and are not intended to limit the scope or practice of the claimed compositions. In addition, the dosage regimen of the compositions of the present disclosure may be based on a number of factors, including the type of disease, the age, weight, sex, medical condition of the subject, the severity of the condition, the route of administration and the particular antibody being used. Thus, dosage regimens may vary widely, but may be routinely determined using standard methods. For example, the dosage may be adjusted based on pharmacokinetic or pharmacodynamic parameters, which may include clinical effects such as toxic effects and/or experimental values. Thus, the present disclosure includes dose escalation (intra-subject dose-delivery) within a subject as determined by the skilled artisan. Determining appropriate dosages and regimens is well known in the relevant art and will be understood to be within the skill of the art once the teachings disclosed herein are provided.
For administration to a human subject, the total monthly dose of the antibodies or antigen-binding fragments thereof of the present disclosure may range from 0.5-1200 mg/subject, 0.5-1100 mg/subject, 0.5-1000 mg/subject, 0.5-900 mg/subject, 0.5-800 mg/subject, 0.5-700 mg/subject, 0.5-600 mg/subject, 0.5-500 mg/subject, 0.5-400 mg/subject, 0.5-300 mg/subject, 0.5-200 mg/subject, 0.5-100 mg/subject, 0.5-50 mg/subject, 1-1200 mg/subject, 1-1100 mg/subject, 1-1000 mg/subject, 1-900 mg/subject, 1-800 mg/subject, 1-700 mg/subject, 1-600 mg/subject, 1-500 mg/subject, 1-400 mg/subject, 1-300 mg/subject, 1-200 mg/subject, 1-100 mg/subject, or 1-50 mg/subject, depending on the mode of administration. For example, a monthly intravenous dose may require about 1-1000 mg/subject. In various embodiments, the antibodies of the present disclosure, or antigen binding fragments thereof, may be administered at about 1-200 mg/subject, 1-150 mg/subject, or 1-100 mg/subject. The total monthly dose may be administered in single or divided doses and may fall outside the typical ranges set forth herein at the discretion of the physician.
Exemplary, non-limiting daily dosing ranges for a therapeutically or prophylactically effective amount of an antibody or antigen binding fragment thereof of the present disclosure may be 0.001mg/kg body weight to 100mg/kg body weight, 0.001mg/kg body weight to 90mg/kg body weight, 0.001mg/kg body weight to 80mg/kg body weight, 0.001mg/kg body weight to 70mg/kg body weight, 0.001mg/kg body weight to 60mg/kg body weight, 0.001mg/kg body weight to 50mg/kg body weight, 0.001mg/kg body weight to 40mg/kg body weight, 0.001mg/kg body weight to 30mg/kg body weight, 0.001mg/kg body weight to 20mg/kg body weight, 0.001mg/kg body weight to 10mg/kg body weight, 0.001mg/kg body weight to 5mg/kg body weight, 0.001mg/kg body weight to 4mg/kg body weight, 0.001mg/kg body weight to 3mg/kg body weight 0.001mg/kg body weight to 2mg/kg body weight, 0.001mg/kg body weight to 1mg/kg body weight, 0.010mg/kg body weight to 50mg/kg body weight, 0.010mg/kg body weight to 40mg/kg body weight, 0.010mg/kg body weight to 30mg/kg body weight, 0.010mg/kg body weight to 20mg/kg body weight, 0.010mg/kg body weight to 10mg/kg body weight, 0.010mg/kg body weight to 5mg/kg body weight, 0.010mg/kg body weight to 4mg/kg body weight, 0.010mg/kg body weight to 3mg/kg body weight, 0.010mg/kg body weight to 2mg/kg body weight, 0.010mg/kg body weight to 1mg/kg body weight, 0.1mg/kg body weight to 50mg/kg body weight, 0.1mg/kg body weight to 40mg/kg body weight, 0.1mg/kg body weight to 30mg/kg body weight, 0.1mg/kg body weight to 20mg/kg body weight, 0.1mg/kg body weight to 10mg/kg body weight, 0.010mg/kg body weight to 10mg/kg body weight, 0.1mg/kg body weight to 5mg/kg body weight, 0.1mg/kg body weight to 4mg/kg body weight, 0.1mg/kg body weight to 3mg/kg body weight, 0.1mg/kg body weight to 2mg/kg body weight, 0.1mg/kg body weight to 1mg/kg body weight, 1mg/kg body weight to 50mg/kg body weight, 1mg/kg body weight to 40mg/kg body weight, 1mg/kg body weight to 30mg/kg body weight, 1mg/kg body weight to 20mg/kg body weight, 1mg/kg body weight to 10mg/kg body weight, 1mg/kg body weight to 5mg/kg body weight, 1mg/kg body weight to 4mg/kg body weight, 1mg/kg body weight to 3mg/kg body weight, 1mg/kg body weight to 2mg/kg body weight, or 1mg/kg body weight to 1mg/kg body weight. It should be noted that the dosage value may vary with the type and severity of the condition to be alleviated. It will also be appreciated that for any particular subject, the particular dosage regimen should be adjusted over time according to the individual needs and the professional judgment of the person administering or supervising the administration of the compositions, and that the dosage ranges set forth herein are exemplary only and are not intended to limit the scope or practice of the claimed compositions.
In various embodiments, the total dose administered will achieve a plasma concentration in the range of, for example, about 1 μg/ml to 1000 μg/ml, about 1 μg/ml to 750 μg/ml, about 1 μg/ml to 500 μg/ml, about 1 μg/ml to 250 μg/ml, about 10 μg/ml to 1000 μg/ml, about 10 μg/ml to 750 μg/ml, about 10 μg/ml to 500 μg/ml, about 10 μg/ml to 250 μg/ml, about 20 μg/ml to 1000 μg/ml, about 20 μg/ml to 750 μg/ml, about 20 μg/ml to 500 μg/ml, about 20 μg/ml to 250 μg/ml, about 30 μg/ml to 1000 μg/ml, about 30 μg/ml to 750 μg/ml, about 30 μg/ml to 500 μg/ml, about 30 μg/ml to 250 μg/ml.
Toxicity and therapeutic index of the pharmaceutical compositions of the invention may be determined by standard pharmaceutical procedures in cell culture or experimental animals, e.g., for determining LD 50 (dose lethal to 50% of the population) and ED 50 (dose therapeutically effective for 50% of the population). The dose ratio between the toxic dose and the therapeutically effective dose is the therapeutic index, and the therapeutic index can be expressed as the ratio LD 50 /ED 50 . Compositions exhibiting large therapeutic indices are generally preferred.
In various embodiments, a single administration or more than one administration of the pharmaceutical composition is administered depending on the dosage and frequency of the subject's needs and tolerability. Regardless, the composition should provide a sufficient amount of at least one antibody or antigen-binding fragment thereof disclosed herein to effectively treat the subject. The dose may be administered once, but may be applied periodically until a therapeutic result is achieved or until the side effect warning ceases therapy.
The frequency of administration of the pharmaceutical composition of the antibody or antigen-binding fragment thereof depends on the nature of the therapy and the particular disease being treated. The subject may be treated at regular intervals, such as weekly or monthly, until the desired therapeutic result is achieved. Exemplary dosing frequencies include, but are not limited to: uninterrupted once a week; weekly every other week; once every 2 weeks; once every 3 weeks; uninterrupted once a week for 2 weeks, then once a month; uninterrupted once a week for 3 weeks, then once a month; once a month; once every two months; once every 3 months; once every 4 months; once every 5 months; or once every 6 months, or once a year.
Combination therapy
As used herein, the terms "co-administration," co-administered, "and" in combination with "in combination with," when referring to an antibody or antigen-binding fragment thereof of the present disclosure and one or more other therapeutic agents, are intended to mean, and include, the following: such a combination of an antibody or antigen-binding fragment thereof of the present disclosure and one or more therapeutic agents is administered simultaneously to a subject in need of treatment, when such components are formulated together in a single dosage form that releases the components to the subject at substantially the same time; such a combination of an antibody or antigen-binding fragment thereof of the present disclosure and one or more therapeutic agents is administered to a subject in need of treatment substantially simultaneously, when such components are formulated separately from each other into separate dosage forms that are taken by the subject at substantially the same time, whereupon the components are released to the subject at substantially the same time; such a combination of an antibody or antigen-binding fragment thereof of the present disclosure and one or more therapeutic agents is administered sequentially to a subject in need of treatment, when such components are formulated separately from each other into separate dosage forms that are taken by the subject at successive times with a significant time interval between each administration, whereupon the components are released to the subject at substantially different times; and such a combination of an antibody or antigen-binding fragment thereof of the present disclosure and one or more therapeutic agents is administered sequentially to a subject in need of treatment, when such components are formulated together into a single dosage form that releases the components in a controlled manner, whereupon they are released simultaneously, sequentially and/or overlapping at the same and/or different times to the subject, wherein each moiety may be administered by the same or different routes.
In another aspect, the invention relates to a combination therapy designed to treat cancer or an infectious disease in a subject, the combination therapy comprising administering to the subject a) a therapeutically effective amount of an isolated antibody or antigen binding fragment of the invention, and b) one or more additional therapies selected from the group consisting of immunotherapy, chemotherapy, small molecule kinase inhibitor targeted therapy, surgery, radiation therapy, vaccination regimen, and stem cell transplantation, wherein the combination therapy provides increased cell killing of tumor cells, i.e., there is a synergy between the isolated antibody or antigen binding fragment and the additional therapies when co-administered.
In various embodiments, the immunotherapy is selected from the group consisting of: treatment with agonistic, antagonistic or blocking antibodies against co-stimulatory or co-inhibitory molecules (immune checkpoints) such as PD-1, PD-L1, OX-40, CD137, GITR, LAG3, TIM-3 and VISTA; use of bispecific T cell engagement antibodiesTreatment such as bordetention; treatment involving administration of biological response modifiers such as IL-2, IL-12, IL-15, IL-21, GM-CSF, IFN- α, IFN- β, and IFN- γ; treatment with therapeutic vaccines such as sipuleucel-T; treatment with dendritic cell vaccines or tumor antigen peptide vaccines; treatment using Chimeric Antigen Receptor (CAR) -T cells; treatment with CAR-NK cells; treatment with Tumor Infiltrating Lymphocytes (TILs); treatment with adoptive transfer of anti-tumor T cells (ex vivo expansion and/or TCR transgene); treatment with TALL-104 cells; and treatment with immunostimulants such as Toll-like receptor (TLR) agonists CpG and imiquimod.
Many conventional compounds have been shown to have antitumor activity. These compounds have been used as agents in chemotherapy to shrink solid tumors, prevent metastasis and further growth, or reduce the number of malignant T cells in leukemia or bone marrow malignancy. Although chemotherapy has been effective in treating many types of malignancies, many antineoplastic compounds induce undesirable side effects. It has been shown that when two or more different treatments are combined, the treatments can work synergistically and allow the dose of each treatment to be reduced, thereby reducing the adverse side effects of each compound at higher doses. In other examples, a malignancy that is refractory to treatment may be responsive to a combination therapy of two or more different treatments.
When the antibodies or antigen-binding fragments disclosed herein are administered in combination, either concomitantly or sequentially, with another conventional anti-neoplastic agent, such antibodies or antigen-binding fragments can enhance the therapeutic effect of the anti-neoplastic agent or overcome the tolerance of the cell to such anti-neoplastic agent. This allows for a reduction in the dosage of the anti-tumor agent, thereby reducing undesirable side effects, or restoring the effectiveness of the anti-tumor agent in tolerogenic T-cells.
For illustration only, pharmaceutical compounds that may be used in combination with anti-tumor therapies include: aminoglutethimide, amsacrine, anastrozole, asparaginase, bcg, bicalutamide, bleomycin, buserelin, busulfan, camptothecine, carboplatin, carmustine, chlorambucil, chlorpyrimide, chlorambucil, and colchicine daunorubicin (daunorubicin), dienestrol (dienestrol), diethylstilbestrol (diethylstilbestrol), docetaxel (docetaxel), doxorubicin (flutoxamycin), gemcitabine (gemcitabine), genistein (genistein), goserelin, hydroxyurea (hydroxyurea), idarubicin (idarubicin), ifosfamide (ifosfamide), imatinib (imatinib), interferon, irinotecan (irinotecan), irinotecan, letrozole (letrozole), leucovorin (leucovorin), leuprorelin (leuprolide), levamisole (levamisole), cyclohexa-nitrourea (lomustine), mechlorethamine (mechlorethamine), medroxyprogesterone (medroxyprogesterone), megestrol (megestrol), melphalan (melphalan), mercaptopurine (mecaptopurine), mesna (mesna), methotrexate (methotrexate), mitomycin (mitomycin), mitotane (mitotane), mitoxantrone (mitoxantrone), nilutamide (nidazole), nocodazole (octreotide), oxaliplatin (oxaliplide), oxaliplatin (paclitaxel), paclitaxel (paclitaxel); pamidronate, penstatin, plicamycin, bufafenamide, thioguanine, thiotepa, titanocene dichloride (titanocene dichloride), topotecan, trastuzumab, retinoic acid (tretamab), vinblastine, neomycin (vincristine), vindesine (vindeline), and vinorelbine (vinorelbine).
These chemotherapeutic anti-tumor compounds can be classified, for example, by the mechanism of their action into the following groups: antimetabolites/anticancer agents such as pyrimidine analogs (5-fluorouracil, fluoroglycoside, capecitabine, gemcitabine and cytarabine) and purine analogs, folic acid antagonists and related inhibitors (mercaptopurine, thioguanine, pravastatin and 2-chlorodeoxyadenosine (cladribine)); antiproliferative/antimitotic agents including natural products such as vinca alkaloids (vinca alkaloids, vincristine and vinorelbine), microtubule interferents such as taxane (taxane) (paclitaxel, docetaxel), vincristine, vinblastine, nocodazole, epothilones (epothilones) and novelin (navilbine), epipodophyllotoxins (etoposide, teniposide), DNA damaging agents (actinomycin, amsacrine, anthracyclines, bleomycin, busulfan, camptothecine, carboplatin, chlorambucil, cisplatin, cyclophosphamide (cytoxan), dactinomycin, daunorubicin, doxorubicin, epirubicin, hexamethylmelamine, oxaliplatin, ifolia, melphalan, mitomycetin, mitoxantrone, nitrosourea, procyanidins, procalcitonin, triotezole (16), trioteh-oxolanide (trioteh), trimethoside (trioteh) and triazamide); antibiotics such as dactinomycin (actinomycin D), daunorubicin, doxorubicin (doxorubicin), idarubicin, anthracyclines, mitoxantrone, bleomycins, plicamycin (mithramycin), and mitomycin; enzymes (L-asparaginase, which metabolizes L-asparagine systemically and deprives cells of the ability to synthesize their own asparagine); antiplatelet agents; antiproliferative/antimitotic alkylating agents such as nitrogen mustards (nitrogen mustards, cyclophosphamide and analogues, melphalan, chlorambucil), ethyleneimines and methyltriamines (hexamethylmelamine and thiotepa), alkyl sulfonate-busulfan (alkyl sulfonates-busulfan), nitrosoureas (carmustine (BCNU) and analogues, streptozotocin), trazenes-Dacarbazinine (DTIC); antiproliferative/antimitotic antimetabolites such as folic acid analogs (methotrexate); platinum coordination complexes (cisplatin, carboplatin), procarbazine, hydroxyurea, mitotane, aminoglutethimide; hormones, hormone analogs (estrogens, tamoxifen, goserelin, bicalutamide, nilutamide) and aromatase inhibitors (letrozole, anastrozole); anticoagulants (heparin, synthetic heparin salts, and other thrombin inhibitors); fibrinolytic agents (fibrinolytic agents) (such as tissue plasminogen activator, streptokinase and urokinase), aspirin, dipyridamole, ticlopidine, clopidogrel, abciximab; an anti-migration agent (antimigratory agent); antisecretory agents (breveldin); immunosuppressants (cyclosporine, tacrolimus (FK-506), sirolimus (rapamycin), azathioprine (azathioprine), mycophenolate mofetil (mycophenolate mofetil)); anti-angiogenic compounds (TNP-470, genistein) and growth factor inhibitors (vascular endothelial growth factor (VEGF) inhibitors, fibroblast Growth Factor (FGF) inhibitors); angiotensin receptor blockers; a nitric oxide donor; an antisense oligonucleotide; antibodies (trastuzumab); cell cycle inhibitors and differentiation inducers (retinoic acid); mTOR inhibitors, topoisomerase inhibitors (doxorubicin), amsacrine, camptothecin, daunorubicin, dactinomycin, eniposide, epirubicin, etoposide, idarubicin and mitoxantrone, topotecan, irinotecan); corticoids (corticosteroid) (cortisone, dexamethasone, hydrocortisone, methylprednisolone, prednisone, and prednisolone); growth factor signal transduction kinase inhibitors; mitochondrial dysfunction inducers and caspase activators; and a chromatin interfering agent.
In various embodiments, the chemotherapy comprises a chemotherapeutic agent selected from the group consisting of: daunorubicin, dactinomycin, doxorubicin, bleomycin, mitomycin, nitrogen mustard, chlorambucil, melphalan, cyclophosphamide, 6-mercaptopurine, 6-thioguanine, bendamustine, cytarabine (CA), 5-fluorouracil (5-FU), fluorouridine (5-FUdR), methotrexate (MTX), colchicine, vincristine, vinblastine, etoposide, teniposide, cisplatin, carboplatin, oxaliplatin, pentostatin, cladribine, cytarabine, gemcitabine, pralatrexate, mitoxantrone, diethylstilbestrol (DES), fludarabine, ifosfamide, hydroxyurea antibiotics (such as paclitaxel and docetaxel), and/or combinations of anthracyclines such as, but not limited to, paclitaxel-EPOCH, CHOP, CVP or FOOX.
In various embodiments, the small molecule kinase inhibitor targeted therapy comprises a small molecule kinase inhibitor selected from the group consisting of: bruton's Tyrosine Kinase (BTK) inhibitors, phosphatidylinositol-3-kinase (PI 3K) inhibitors, SYK inhibitors (e.g., entospletinib), AKT inhibitors, mTOR inhibitors, src inhibitors, JAK/STAT inhibitors, ras/Raf/MEK/ERK inhibitors, and Aurora inhibitors (see D' Cruz et al, expert Opin Pharmacother,14 (6): 707-21, 2013).
In various embodiments, the combination therapy comprises simultaneous administration of the antibody or antigen-binding fragment thereof and one or more additional therapies. In various embodiments, the antibody or antigen-binding fragment composition thereof and the one or more additional therapies are administered sequentially, i.e., the antibody or antigen-binding fragment composition thereof is administered before or after the administration of the one or more additional therapies.
In various embodiments, the administration of the antibody or antigen binding fragment composition thereof and the one or more additional therapies is simultaneous, i.e., the administration periods of the antibody or antigen binding fragment composition thereof and the one or more additional therapies overlap each other.
In various embodiments, the administration of the antibody or antigen binding fragment composition thereof and one or more additional therapies is not concurrent. For example, in various embodiments, administration of the antibody or antigen binding fragment composition thereof is terminated prior to administration of the one or more additional therapies. In various embodiments, administration of one or more additional therapies is terminated prior to administration of the antibody or antigen binding fragment composition thereof.
When an antibody or antigen-binding fragment thereof disclosed herein is administered concomitantly or sequentially in combination with one or more additional therapies, such an antibody or antigen-binding fragment thereof may enhance the therapeutic effect of or overcome the tolerance of the cell to the one or more additional therapies. This allows for a reduction in the dosage of or a reduction in the duration of one or more additional therapies, thereby reducing undesired side effects, or restoring the efficacy of one or more additional therapies.
Diagnostic use
In another aspect, the invention provides methods for detecting the presence of a human PD-L1 antigen in a sample in vitro or in vivo, e.g., for diagnosing a human PD-L1-related disorder. In some methods, this is accomplished by contacting the sample to be tested as well as the control sample with a human sequence antibody or a human monoclonal antibody or antigen binding portion thereof (or a bispecific molecule or a multispecific molecule) of the invention under conditions that allow for the formation of a complex between the antibody and human PD-L1. Complex formation is then detected (e.g., using ELISA) in both samples, and any statistically significant difference in complex formation between the samples is indicative of the presence of human PD-L1 antigen in the test sample.
In various embodiments, methods for detecting cancer or determining a diagnosis of cancer in a subject are provided. The method comprises contacting a biological sample from a subject with an isolated antibody or antigen-binding fragment thereof of the invention, and detecting binding of the isolated human monoclonal antibody or antigen-binding fragment thereof to the sample. An increase in binding of the isolated human monoclonal antibody or antigen binding fragment thereof to the sample as compared to binding of the isolated human monoclonal antibody or antigen binding fragment thereof to the control sample detects or determines a diagnosis of cancer in the subject. The control may be a sample from a subject known not to have cancer, or a standard value. The sample may be any sample including, but not limited to, tissue from biopsies, autopsies and pathological specimens. Biological samples also include tissue sections, e.g., frozen sections taken for histological purposes. Biological samples also include body fluids such as blood, serum, plasma, sputum, and spinal fluid.
In one embodiment, a kit for detecting PD-L1 in a biological sample (such as a blood sample) is provided. Kits for detecting polypeptides will typically comprise a human antibody that specifically binds to PD-L1, such as any of the antibodies disclosed herein. In some embodiments, antibody fragments, such as Fv fragments, are included in a kit. For in vivo use, the antibody may be an scFv fragment. In further embodiments, the antibody is labeled (e.g., with a fluorescent, radioactive, or enzymatic label).
In one embodiment, the kit includes instructional materials disclosing means for using antibodies that specifically bind to PD-L1. The instructional material may be written in electronic form, such as a computer diskette or optical disk, or may be visual, such as a video file. The kit may also include additional components that facilitate the particular application for which the kit is designed. Thus, for example, the kit may additionally comprise means (means) for detecting the label (such as an enzyme substrate for an enzyme label, a filter set for detecting a fluorescent label, a suitable secondary label such as a secondary antibody, etc.). The kit may additionally contain buffers and other reagents conventionally used to practice a particular method. Such kits and appropriate components are well known to those skilled in the art.
In one embodiment, the diagnostic kit comprises an immunoassay. Although the details of the immunoassay may vary with the particular format used, methods of detecting PD-L1 in a biological sample generally include the step of contacting the biological sample with an antibody that specifically reacts with PD-L1 under immunological reaction conditions. Antibodies are allowed to specifically bind under immunological reaction conditions to form immune complexes, and the presence of immune complexes (bound antibodies) is detected directly or indirectly.
In various embodiments, the antibody or antigen binding fragment may or may not be labeled for diagnostic purposes. In general, diagnostic assays require detection of complex formation resulting from binding of antibodies to PD-L1. The antibody may be directly labeled. A number of labels may be used including, but not limited to, radionuclides, fluorescers (fluorscers), enzymes, enzyme substrates, enzyme cofactors, enzyme inhibitors, and ligands (e.g., biotin, haptens). Many suitable immunoassays are known to the skilled artisan (see, for example, U.S. Pat. Nos. 3,817,827; 3,850,752; 3,901,654; and 4,098,876). When unlabeled, the antibodies can be used in assays, such as agglutination assays. Unlabeled antibodies may also be used in combination with additional suitable reagent(s) that may be used to detect antibodies, such as labeled antibodies (e.g., secondary antibodies) or other suitable reagents (e.g., labeled protein a) that react with a primary antibody (e.g., an anti-idiotype antibody or other antibody specific for unlabeled immunoglobulin).
The antibodies or antigen binding fragments provided herein may also be used in methods of detecting a susceptibility of a mammal to certain diseases. To illustrate, the method can be used to detect a susceptibility of a mammal to a disease based on the amount of PD-L1 present on the cells and/or the progression of the number of PD-L1 positive cells in the mammal. In one embodiment, the present application provides a method of detecting a susceptibility of a mammal to a tumor. In this embodiment, the sample to be tested is contacted with an antibody that binds to PD-L1 or a portion thereof under conditions suitable for the antibody to bind thereto, wherein the sample comprises cells that express PD-L1 in a normal individual. Detecting binding of the antibody and/or the amount of binding, which is indicative of the individual's susceptibility to the tumor, wherein a higher level of the receptor correlates with an increased susceptibility of the individual to the tumor.
In various embodiments, the antibody or antigen binding fragment is attached to a label that can be detected (e.g., the label can be a radioisotope, a fluorescent compound, an enzyme, or an enzyme cofactor). The active moiety may be a radioactive agent, such as: positron emitters of radioactive heavy metals such as iron chelates, radioactive chelates of gadolinium or manganese, oxygen, nitrogen, iron, carbon or gallium, 43 K、 52 Fe、 57 Co、 67 Cu、 67 Ga、 68 Ga、 123 I、 125 I、 131 I、 132 I or 99 Tc. Binding agents attached to such moieties can be used as imaging agents and administered in amounts effective for diagnostic use in mammals such as humans, and then the localization and accumulation of the imaging agents is detected. The positioning and accumulation of the imaging agent can be accomplished by radioscintigraphy,Nuclear magnetic resonance imaging, computed tomography or positron emission tomography.
Immunoscintigraphy using antibodies or antigen binding fragments directed against PD-L1 can be used to detect and/or diagnose cancer and vascular systems. For example, for 99 Technetium, technetium, 111 Indium or indium 125 Monoclonal antibodies to iodine-labeled PD-L1 markers can be effectively used for such imaging. As will be apparent to those skilled in the art, the amount of radioisotope to be administered depends on the radioisotope. One of ordinary skill in the art can readily formulate the amount of imaging agent to be administered based on the specific activity and energy of a given radionuclide used as the active moiety. Typically, 0.1 to 100 millicuries, or 1 to 10 millicuries, or 2 to 5 millicuries, are administered per dose of imaging agent. Thus, the disclosed compositions are useful as imaging agents comprising a targeting moiety conjugated to a radioactive moiety comprising 0.1 to 100 millicuries, in some embodiments 1 to 10 millicuries, in some embodiments 2 to 5 millicuries, and in some embodiments 1 to 5 millicuries.
Immunoconjugates
The present application also provides immunoconjugates comprising an antibody of the invention, or an antigen-binding fragment thereof, conjugated (or linked) directly or indirectly to an effector molecule. In this aspect, the term "conjugated" or "linked" refers to the formation of two polypeptides into one continuous polypeptide molecule. The ligation may be performed by chemical means or recombinant means. In one embodiment, the linkage is chemical, wherein the reaction between the antibody moiety and the effector molecule has produced a covalent bond formed between the two molecules to form one molecule. Peptide linkers (short peptide sequences) may optionally be included between the antibody and the effector molecule. In various embodiments, the antibody or antigen binding fragment is linked to an effector molecule. In other embodiments, the antibody or antigen binding fragment linked to the effector molecule is further linked to a lipid, protein, or peptide to increase its half-life in vivo. Thus, in various embodiments, the antibodies of the present disclosure can be used to deliver a variety of effector molecules.
The effector molecule may be a detectable label, an immunotoxin, a cytokine, a chemokine, a therapeutic agent, or a chemotherapeutic agent.
Specific, non-limiting examples of immunotoxins include, but are not limited to, abrin (abrin), ricin (ricin), pseudomonas exotoxin (Pseudomonas exotoxin) (PE such as PE35, PE37, PE38, and PE 40), diphtheria Toxin (DT), botulinum toxin (botulinum toxin), cholix toxin, or modified toxins thereof, or other toxic agents that directly or indirectly inhibit cell growth or kill cells.
A "cytokine" is a class of proteins or peptides released by one cell population that act as intercellular mediators on another cell. Cytokines may act as immunomodulators. Examples of cytokines include lymphokines, monokines, growth factors, and traditional polypeptide hormones. Thus, embodiments may utilize interferons (e.g., IFN- α, IFN- β, and IFN- γ); tumor Necrosis Factor Superfamily (TNFSF) members; human growth hormone; thyroxine; insulin; pre-insulin; relaxin; pre-relaxin; follicle Stimulating Hormone (FSH); thyroid Stimulating Hormone (TSH); luteinizing Hormone (LH); liver growth factors; prostaglandin, fibroblast growth factor; prolactin; placental lactation hormone, OB protein; TNF-alpha; TNF-beta; an integrin; thrombopoietin (TPO); nerve growth factors such as NGF- β; platelet growth factors; TGF-alpha; TGF- β; insulin-like growth factor-I and insulin-like growth factor-II; erythropoietin (EPO); colony Stimulating Factors (CSF) such as macrophage-CSF (M-CSF); granulocyte-macrophage-CSF (GM-CSF); and granulocyte-CSF (G-CSF); interleukins (IL-1 to IL-21), kit-ligands or FLT-3, angiostatin, thrombospondin or endostatin. These cytokines include proteins from natural sources or from recombinant cell cultures and biologically active equivalents of the native sequence cytokines.
Chemokines may also be conjugated to antibodies disclosed herein. Chemokines are a small (approximately about 4kDa to about 14 kDa), superfamily of inducible and secreted pro-inflammatory cytokines that act primarily as chemoattractants and activators of specific leukocyte subtypes. Chemokine production is induced by inflammatory cytokines, growth factors and pathogenic stimuli. Chemokine proteins are divided into subfamilies (α, β, and δ) based on conserved amino acid sequence motifs, and are classified into four highly conserved groups, CXC, CC, C, and CX3C, based on the position of the first two cysteines adjacent to the amino terminus. To date, more than 50 chemokines have been found, and at least 18 human seven transmembrane domain (7 TM) chemokine receptors are present. Chemokines used include, but are not limited to RANTES, MCAF, MCP-1 and fractalkine.
The therapeutic agent may be a chemotherapeutic agent. The chemotherapeutic agents used can be readily identified by those skilled in the art (see, for example, harrison, chapter Principles of Internal Medicine, 14 th edition, chapter 86, slapak and Kufe, principles of Cancer Therapy; perry et al, abeloff, chapter Clinical Oncology. Sup. Nd. Chapter 17, chemothepy. COPYRIGHT.2000 Churchill Livingstone, inc; baltzer L., berkery R. (eds.): oncology Pocket Guide to Chemotherapy, second edition St. Louis, mosby-Year Book,1995;Fischer D S,Knobf M F,Durivage H J (eds.): the Cancer Chemotherapy Handbook, 4 th edition, st. Louis, mosby-Year Book, 1993). Useful chemotherapeutic agents for preparing immunoconjugates include auristatin, dolastatin, MMAE, MMAF, AFP, DM, AEB, doxorubicin, daunorubicin, methotrexate, melphalan, chlorambucil, vinca alkaloids, 5-fluorouridine, mitomycin-C, paclitaxel, L-asparaginase, mercaptopurine, thioguanine, hydroxyurea, cytarabine, cyclophosphamide, ifosfamide, nitrosoureas, cisplatin, carboplatin, mitomycin, azotemamide, procarbazine, topotecan, nitrogen mustards, cyclophosphamide (cytoxan), etoposide, BCNU, irinotecan, camptothecine, bleomycin, idarubicin, dactinomycin, procyanidins, mitoxantrone, asparaginase, vincristine, vinorelbine, paclitaxel, and docetaxel, and salts, solvates or derivatives thereof. In various embodiments, the chemotherapeutic agent is auristatin E (also known in the art as dolastatin-10) or a derivative thereof, as well as a pharmaceutically salt or solvate thereof. Typical auristatin derivatives include DM1, AEB, AEVB, AFP, MMAF and MMAE. auristatin E and its derivatives, synthesis and structure of linkers are described in, for example, U.S. patent application publication No. 20030083263; U.S. patent application publication No. 20050238629; and U.S. patent No. 6,884,869 (each of which is incorporated herein by reference in its entirety). In various embodiments, the therapeutic agent is an auristatin or an auristatin derivative. In various embodiments, the auristatin derivative is doraline-valine-dolaseline-dolaplatin-phenylalanine (MMAF) or monomethyauristatin E (MMAE). In various embodiments, the therapeutic agent is maytansine (maytansinoid) or a maytansinol (maytansinol) analog. In various embodiments, the maytansinoid is DM1.
The effector molecule may be attached to the antibody or antigen binding fragment of the invention using any number of means known to those skilled in the art. Both covalent and non-covalent attachment means may be used. The procedure for attaching the effector molecule to the antibody varies depending on the chemical structure of the effector molecule. Polypeptides typically comprise a variety of functional groups; such as carboxylic acids (COOH), free amines (- -NH) 2 ) Or a thiol (- -SH) group, which can be used to react with a suitable functional group on an antibody to cause binding of an effector molecule. Optionally, the antibody is derivatized to expose or attach additional reactive functional groups. Derivatization may include attachment of any of a number of linker molecules, such as those available from Pierce Chemical Company, rockford, III. The linker may be any molecule used to attach the antibody to the effector molecule. The linker is capable of forming a covalent bond with both the antibody and with the effector molecule. Suitable linkers are well known to those skilled in the art and include, but are not limited to, straight or branched chain carbon linkers, heterocyclic carbon linkers, or peptide linkers. Where the antibody and effector molecule are polypeptides, the linker may be attached to the constitutive amino acid or to the alpha carbon amino and carboxyl groups of the attached terminal amino acid through its side chain groups (such as through disulfide linkages to cysteines).
In some cases, it is desirable that the effector molecule be free from the antibody when the immunoconjugate has reached its target site. Thus, in these cases, the immunoconjugate will comprise a linkage that is cleavable near the target site. Cleavage of the linker to release the effector molecule from the antibody may be facilitated by the enzymatic activity or conditions experienced by the immunoconjugate in the target cell or in the vicinity of the target site.
Procedures for conjugating antibodies to effector molecules have been previously described and are within the purview of those skilled in the art. For example, procedures for preparing enzymatically active polypeptides of immunotoxins are described in WO84/03508 and WO85/03508, which are incorporated herein by reference for the purpose of their specific teachings. Other techniques are described in Shih et al, int.J. cancer 41:832-839 (1988); shih et al, int.J.cancer 46:1101-1106 (1990); shih et al, U.S. Pat. nos. 5,057,313; shih Cancer Res.51:4192, international publication WO 02/088172; U.S. patent No. 6,884,869; international patent publication WO2005/081711; U.S. published application 2003-013089A; and U.S. patent application No. 2008055044, each of which is incorporated herein by reference for the purpose of teaching such techniques.
The immunoconjugates of the invention retain the immunoreactivity of the antibody or antigen-binding fragment, e.g., the antibody or antigen-binding fragment has approximately the same, or only slightly reduced, ability to bind antigen after conjugation as before conjugation. As used herein, immunoconjugates are also referred to as Antibody Drug Conjugates (ADCs).
Bispecific molecules
In another aspect, the invention features a bispecific molecule comprising an anti-PD-L1 antibody or antigen-binding fragment thereof of the invention. The antibodies or antigen binding fragments thereof of the invention may be derivatized or linked to another functional molecule, such as another peptide or protein (e.g., another antibody or ligand for a receptor), to generate bispecific molecules that bind to at least two different binding sites or target molecules. The antibodies of the invention may in fact be derivatized or linked to more than one other functional molecule to generate multispecific molecules that bind to more than two different binding sites and/or target molecules; such multispecific molecules are also intended to be encompassed by the term "bispecific molecule" as used herein. To produce the bispecific molecules of the invention, the antibodies of the invention may be functionally linked (e.g., by chemical coupling, genetic fusion, non-covalent association, or other means) to one or more other binding molecules, such as another antibody, antibody fragment, peptide, or binding mimetic, such that the bispecific molecule is produced. In various embodiments, the invention includes bispecific molecules capable of binding to both effector cells (e.g., monocytes, macrophages or polymorphonuclear cells (PMNs)) expressing fcγr or fcαr and to target cells expressing PD. In such embodiments, the bispecific molecule targets the PD-L1 expressing cell to an effector cell and triggers Fc receptor mediated effector cell activity, e.g., phagocytosis, antibody dependent cell mediated cytotoxicity (ADCC), cytokine release, or the generation of superoxide anions, of the PD-L1 expressing cell. Methods for preparing bispecific molecules of the invention are well known in the art.
Polynucleotide and antibody expression
The present application also provides polynucleotides comprising nucleotide sequences encoding anti-PD-L1 antibodies or antigen-binding fragments thereof. Because of the degeneracy of the genetic code, many nucleic acid sequences encode an antibody amino acid sequence. The present application also provides polynucleotides that hybridize to polynucleotides encoding antibodies that bind to human PD-L1 under stringent hybridization conditions or lower stringent hybridization conditions, e.g., as defined herein.
Stringent hybridization conditions include, but are not limited to, hybridization with filter-bound DNA in 6 XSSC at about 45℃followed by one or more washes in 0.2 XSSC/0.1% SDS at about 50-65℃and highly stringent conditions such as hybridization with filter-bound DNA in 6 XSSC at about 45℃followed by one or more washes in 0.1 XSSC/0.2% SDS at about 60℃or any other stringent hybridization conditions known to those of skill in the art (see, e.g., ausubel, F.M. Et al, eds.1989 Current Protocols in Molecular Biology, vol.1, green Publishing Associates, inc. and John Wiley and Sons, inc., NY, pages 6.3.1 through 6.3.6 and 2.10.3).
The polynucleotides may be obtained and the nucleotide sequence of the polynucleotides determined by any method known in the art. For example, if the nucleotide sequence of the antibody is known, the polynucleotide encoding the antibody may be assembled from chemically synthesized oligonucleotides (e.g., as described in Kutmeier et al, bioTechiques 17:242 (1994)), which in brief, involves synthesizing overlapping oligonucleotides comprising portions of the antibody-encoding sequence, annealing and ligating the oligonucleotides, and then amplifying the ligated oligonucleotides by PCR. In one embodiment, codons used include those that are typical for humans or mice (see, e.g., nakamura, Y., nucleic Acids Res.28:292 (2000)).
Polynucleotides encoding antibodies may also be generated from nucleic acids from suitable sources. If a clone comprising a nucleic acid encoding a particular antibody is not available, but the sequence of the antibody molecule is known, then the nucleic acid encoding the immunoglobulin may be chemically synthesized or obtained from a suitable source (e.g., an antibody cDNA library, or a cDNA library generated from any tissue or cell expressing the antibody, or a nucleic acid isolated from any tissue or cell expressing the antibody, preferably polyA+RNA, such as a hybridoma cell selected to express the antibody) by PCR amplification using synthetic primers hybridizable to the 3 'and 5' ends of the sequence, or by cloning using oligonucleotide probes specific for the particular gene sequence to identify cDNA clones, e.g., from a cDNA library encoding the antibody. The amplified nucleic acid produced by PCR can then be cloned into a replicable cloning vector by any method well known in the art.
The invention also relates to host cells expressing the PD-L1 polypeptides and/or anti-PD-L1 antibodies of the invention. A wide variety of host expression systems known in the art can be used to express the antibodies of the invention, including prokaryotic (bacterial) and eukaryotic expression systems, such as yeast, baculovirus (baculovirus), plant, mammalian and other animal cells, transgenic animals and hybridoma cells, and phage display expression systems.
Antibodies of the invention can be prepared by recombinant expression of immunoglobulin light and heavy chain genes in host cells. To recombinantly express an antibody, a host cell is transformed, transduced, infected, etc., with one or more recombinant expression vectors carrying DNA fragments encoding immunoglobulin light and/or heavy chains of the antibody such that the light and/or heavy chains are expressed in the host cell. The heavy and light chains may be independently expressed from different promoters to which they are operably linked in one vector, or alternatively, the heavy and light chains may be independently expressed from different promoters operably linked in two vectors, one vector expressing the heavy chain and one vector expressing the light chain. Optionally, the heavy and light chains may be expressed in different host cells.
Alternatively, the recombinant expression vector may encode a signal peptide that facilitates secretion of the antibody light and/or heavy chain from the host cell. The antibody light chain and/or heavy chain genes may be cloned into a vector such that the signal peptide is operably linked in-frame to the amino terminus of the antibody chain gene. The signal peptide may be an immunoglobulin signal peptide or a heterologous signal peptide. Preferably, the recombinant antibody is secreted into the medium in which the host cells are cultured, from which the antibody can be recovered or purified.
The isolated DNA encoding the HCVR may be converted to a full length heavy chain gene by operably linking the DNA encoding the HCVR to another DNA molecule encoding a heavy chain constant region. The sequences of human and other mammalian heavy chain constant region genes are known in the art. DNA fragments comprising these regions may be obtained by, for example, standard PCR amplification. The heavy chain constant region can be of any type (e.g., igG, igA, igE, igM or IgD), class (e.g., igG) 1 、IgG 2 、IgG 3 And IgG 4 ) Or a subclass constant region and any variant thereof, as described in Kabat (above).
The isolated DNA encoding the LCVR region can be converted to a full length light chain gene (and to a Fab light chain gene) by operably linking the DNA encoding the LCVR to another DNA molecule encoding a light chain constant region. The sequences of human and other mammalian light chain constant region genes are known in the art. DNA fragments containing these regions can be obtained by standard PCR amplification. The light chain constant region may be a kappa or lambda constant region.
In addition to one or more antibody heavy and/or light chain genes, the recombinant expression vectors of the invention carry regulatory sequences that control the expression of the one or more antibody chain genes in a host cell. The term "regulatory sequence" is intended to include promoters, enhancers and other expression control elements (e.g., polyadenylation signals) that control the transcription or translation of one or more antibody chain genes, as desired. The design of the expression vector, including the selection of regulatory sequences, may depend on factors such as the choice of host cell to be transformed, the level of expression of the desired protein, and the like. Preferred regulatory sequences for mammalian host cell expression include viral elements that direct high levels of protein expression in mammalian cells, such as promoters and/or enhancers derived from Cytomegalovirus (CMV), simian virus 40 (SV 40), adenoviruses (e.g., adenovirus major late promoter (AdMLP)), and/or polyomaviruses.
Additionally, the recombinant expression vectors of the invention may carry additional sequences, such as sequences that regulate replication of the vector in a host cell (e.g., an origin of replication) and one or more selectable marker genes. Selectable marker genes facilitate selection of host cells into which the vector has been introduced. For example, in general, selectable marker genes confer resistance to drugs such as G418, hygromycin or methotrexate on host cells into which the vector has been introduced. Preferred selectable marker genes include the dihydrofolate reductase (dhfr) gene (for use in dhfr negative host cells selected/amplified with methotrexate), the neo gene (for G418 selection), and GS in Glutamine Synthetase (GS) negative cell lines for selection/amplification, such as NSO.
To express the light and/or heavy chains, one or more expression vectors encoding the heavy and/or light chains are introduced into the host cell by standard techniques, such as electroporation, calcium phosphate precipitation, DEAE-dextran transfection, transduction, infection, and the like. Although expression of the antibodies of the invention in a prokaryotic or eukaryotic host cell is theoretically possible, eukaryotic cells are preferred, and mammalian host cells are most preferred, as such cells are more likely to assemble and secrete properly folded and immunologically active antibodies. Preferred mammalian host cells for expression of the recombinant antibodies of the invention include chinese hamster ovary (CHO cells) [ including DHFR-negative CHO cells, as described by Urlaub and Chasin, proc. Natl. Acad. Sci. USA 77:4216-20,1980, for use with DHFR selectable markers, as described by Kaufman and Sharp, j. Mol. Biol.159:601-21,1982 ], NSO myeloma cells, COS cells and SP2/0 cells. When a recombinant expression vector encoding an antibody gene is introduced into a mammalian host cell, the antibody is produced by culturing the host cell for a period of time sufficient to allow expression of the antibody in the host cell, or more preferably, secreting the antibody into a medium in which the host cell is grown under appropriate conditions known in the art. Antibodies can be recovered from host cells and/or culture medium using standard purification methods.
The present invention provides host cells comprising a nucleic acid molecule according to the invention. Preferably, the host cell of the invention comprises one or more vectors or constructs comprising the nucleic acid molecules of the invention. For example, a host cell of the invention is a cell into which has been introduced a vector of the invention comprising a polynucleotide encoding an LCVR of an antibody of the invention and/or a polynucleotide encoding an HCVR of the invention. The invention also provides a host cell into which the two vectors of the invention have been introduced; a vector comprises a polynucleotide encoding an LCVR of an antibody of the invention, and a vector comprises a polynucleotide encoding an HCVR present in an antibody of the invention, and each polynucleotide is operably linked to an enhancer/promoter regulatory element (e.g., derived from SV40, CMV, adenovirus, etc., such as a CMV enhancer/AdMLP promoter regulatory element or an SV40 enhancer/AdMLP promoter regulatory element) to drive high level transcription of a gene.
After expression, the intact antibodies, individual light and heavy chains, or other immunoglobulin forms of the invention may be purified according to standard procedures in the art, including ammonium sulfate precipitation, ion exchange, affinity (e.g., protein a), reverse phase, hydrophobic interaction column chromatography, hydroxyapatite chromatography, gel electrophoresis, and the like. Standard procedures for purification of therapeutic antibodies are described, for example, by Feng L1, joe x.zhou, xiaoming Yang, tim Tressel and Brian Lee in the article entitled "Current Therapeutic Antibody Production and Process Optimization" (BioProcessing Journal, 9/10 month 2005), incorporated by reference in its entirety for the purpose of teaching purification of therapeutic antibodies. In addition, standard techniques for removing viruses from recombinantly expressed antibody preparations are known in the art (see, e.g., gerd Kern and Mani Krishnan, "Viral Removal by Filtration: points to Consider" (Biopharm International, 10 2006)). It is known that the effectiveness of filtration to remove virus from a formulation of therapeutic antibodies depends at least in part on the concentration of protein and/or antibody of the solution to be filtered. The purification process for the antibodies of the invention may include a step of filtration to remove virus from the mainstream of one or more chromatographic procedures. Preferably, the chromatographic main stream comprising the antibodies of the invention is diluted or concentrated to give a total protein concentration and/or total antibody concentration of about 1g/L to about 3g/L prior to filtration through a pharmaceutical grade nanofiltration for virus removal. Even more preferably, the nanofilter is a DV20 nanofilter (e.g., pall Corporation; east Hills, N.Y.). Substantially pure immunoglobulins of at least about 90%, about 92%, about 94% or about 96% homogeneity, and most preferably about 98% to about 99% or more homogeneity, are preferred for pharmaceutical use. Once partially purified or purified to homogeneity as desired, the sterile antibodies can then be used therapeutically as directed herein.
In view of the discussion above, the invention also relates to an antibody obtainable by a method comprising the step of culturing a host cell comprising, but not limited to, a mammal, plant, bacterium, transgenic animal or transgenic plant cell that has been transformed with a polynucleotide or vector comprising a nucleic acid molecule encoding an antibody of the invention such that the nucleic acid is expressed and optionally recovering the antibody from the host cell culture medium.
In certain aspects, the present application provides hybridoma cell lines, as well as monoclonal antibodies produced by these hybridoma cell lines. The disclosed cell lines have other uses in addition to for the production of monoclonal antibodies. For example, the cell line may be fused with other cells (such as suitably drug-labeled human myeloma, mouse myeloma, human-mouse hybrid myeloma, or human lymphoblastic cells) to produce additional hybridomas, and thus provide for the transfer of genes encoding monoclonal antibodies. In addition, cell lines can be used as a source of nucleic acid encoding an anti-PD-L1 immunoglobulin chain, which can be isolated and expressed (e.g., when transferred to other cells using any suitable technique) (see, e.g., cabily et al, U.S. Pat. No. 4,816,567; winter, U.S. Pat. No. 5,225,539)). For example, clones containing rearranged anti-PD-L1 light or heavy chains may be isolated (e.g., by PCR), or a cDNA library may be prepared from mRNA isolated from the cell line, and cDNA clones encoding anti-PD-L1 immunoglobulin chains may be isolated. Thus, nucleic acids encoding the heavy and/or light chains of an antibody or portions thereof can be obtained and used according to recombinant DNA techniques for producing specific immunoglobulins, immunoglobulin chains, or variants thereof (e.g., humanized immunoglobulins) in a variety of host T cells or in an in vitro translation system. For example, a nucleic acid, including a cDNA or derivative thereof, encoding a variant, such as a humanized immunoglobulin or immunoglobulin chain, may be placed into a suitable prokaryotic or eukaryotic vector (e.g., an expression vector) and introduced into a suitable host T cell by a suitable method (e.g., transformation, transfection, electroporation, infection) such that the nucleic acid is operably linked to one or more expression control elements (e.g., in the vector or integrated into the host T cell genome). For production, the host T cells may be maintained under conditions suitable for expression (e.g., in the presence of an inducer, a suitable medium supplemented with appropriate salts, growth factors, antibiotics, nutritional supplements, etc.), thereby producing the encoded polypeptide. The encoded protein may be recovered and/or isolated (e.g., from the host T cell or culture medium), if desired. It will be appreciated that the method of production involves expression in host T cells of the transgenic animal (see, for example, WO 92/03918 of GenPharm International published 3/19 1992) (incorporated by reference in its entirety).
The host cell may also be used to produce portions or fragments of whole antibodies, such as Fab fragments or scFv molecules, by conventional techniques. For example, it may be desirable to transfect a host cell with DNA encoding the light or heavy chain of an antibody of the invention. Recombinant DNA techniques can also be used to remove some or all DNA encoding one or both of the light and heavy chains that are not necessary for binding to human PD-L1. Antibodies of the invention also include molecules expressed from such truncated DNA molecules.
Methods for expressing single chain antibodies from bacteria such as E.coli (E.coli) and/or refolding into suitable active forms, including single chain antibodies, have been described and are well known and suitable for use with the antibodies disclosed herein (see, e.g., buchner et al, anal biochem.205:263-270,1992;Pluckthun,Biotechnology 9:545,1991;Huse et al, science 246:1275,1989 and Ward et al, nature 341:544,1989, all incorporated herein by reference).
Typically, a functional heterologous protein from E.coli or other bacteria is isolated from inclusion bodies and needs to be solubilized using a strong denaturing agent and subsequently refolded. During the solubilization step, a reducing agent must be present to separate the disulfide bonds, as is well known in the art. Exemplary buffers with reducing agents are: 0.1M Tris pH 8, 6M guanidine, 2mM EDTA, 0.3M DTE (dithioerythritol). Reoxidation of disulfide bonds may occur in the presence of low molecular weight thiol reagents in reduced and oxidized form, as described in Saxena et al, biochemistry 9:5015-5021,1970 (incorporated herein by reference), and in particular as described by Buchner et al, supra.
Renaturation is typically achieved by diluting (e.g., 100-fold) the denatured and reduced protein into refolding buffer. Exemplary buffers are 0.1M Tris, pH 8.0, 0.5M L-arginine, 8mM oxidized glutathione (GSSG) and 2mM EDTA.
As a modification to the diabody purification scheme, the heavy and light chain regions are solubilized and reduced separately and then combined in refolding solution. Exemplary yields are obtained when the two proteins are mixed in a molar ratio such that one protein does not exceed a 5-fold molar excess over the other protein. After redox reorganization is completed, excess oxidized glutathione or other oxidized low molecular weight compounds may be added to the refolding solution.
In addition to recombinant methods, the antibodies, labeled antibodies, and antigen-binding fragments thereof disclosed herein may also be constructed in whole or in part using standard peptide synthesis. Solid phase synthesis of polypeptides of less than about 50 amino acids in length can be accomplished by attaching the C-terminal amino acid of the sequence to an insoluble support, followed by sequential addition of the remaining amino acids in the sequence. The techniques used for solid phase Synthesis consist of Barany & Merrifield, the Peptides: analysis, synthesis, biology, volume 2: special Methods in Peptide Synthesis, section A, pages 3-284; merrifield et al, J.Am.chem.Soc.85:2149-2156,1963 and Stewart et al, solid Phase Peptide Synthesis, 2 nd edition, pierce chem.Co., rockford, ill., 1984. Proteins of greater length can be synthesized by condensation of the amino-and carboxyl-termini of shorter fragments. Methods for forming peptide bonds by activation of the carboxyl terminus, such as by using the coupling agent N, N' -dicyclohexylcarbodiimide, are well known in the art.
The following examples are provided to more fully illustrate the invention, but should not be construed as limiting the scope of the invention.
Example 1
Generation of monoclonal antibodies specifically targeting human PD-L1
Construction of a cell having human IgG1 (Pro) at the C-terminus 100 -Lys 330 ) Tagged human PD-L1 (NP-054862.1, phe) 19 -Thr 239 ) (designated PDL1/Fc fusion protein), expressed in the mouse myeloma cell line (NSO), purified by a protein a column and used as an immunogen. The estimated purity of PDL1/Fc was higher than 90% based on SDS-PAGE under reducing conditions and visualized by silver staining.
Female Balb/C and C57Bl/6 mice were immunized three times (once every two weeks) with 50 μg PDL 1/Fc/mouse intraperitoneally (i.p.). At the first immunization, the antigen was injected as a 1:1 mixture with complete Freund's adjuvant (Sigma, st. Louis, MO) and in the second and third doses as a 1:1 mixture with incomplete Freund's adjuvant (Sigma, st. Louis, MO). Mice were given final boost with 25 μg PDL1/Fc by tail vein injection, and spleen cells were harvested 4 days later for fusion with myeloma cell line NS0 from ATCC (Allendale, NJ). Electrofusion methods were used to obtain hybridoma cells, and hybridoma supernatants were then screened for antigen binding, ligand blocking, igG analysis (binding), reference antibody binding, and FACS binding. 16 mabs were selected from the initial binder screen for subcloning (limiting dilution method) and further evaluation. BD cell MAb medium was used to grow hybridomas in roller bottles for collection of supernatant for antibody production. The mAb was purified by protein a affinity chromatography. The estimated purity of mAb was higher than 90% based on SDS-PAGE coomassie staining. Secondary screening of 12 purified mabs included: PD-L1 binding assay (ELISA), murine PD-L1 binding assay (ELISA), PD1/PD-1 ligand blocking assay by FACS and epitope analysis (epitope binding) screening. Three mabs showed mouse cross-reactivity and epitope analysis (epitope binding) identified 3 epitopes.
Based on the cumulative results of the secondary assays, purified mabs pdl1#1 and pdl1#10 were selected for sequencing and further analysis. FollowTechnical manual for reagents total RNA was extracted from frozen hybridoma cells. Total RNA was analyzed by agarose gel electrophoresis. Following PrimeScript TM Technical manual for first strand cDNA synthesis kit total RNA was reverse transcribed into cDNA using isotype specific antisense primers or universal primers. PCR is then performed to amplify the variable regions (heavy and light chains) of the antibody, which are then individually cloned into standard cloning vectors and sequenced. PDA1#1 and PDL1#10 contain the heavy chain variable region sequences set forth in SEQ ID NOS 12 and 14, respectively, and contain the heavy chain variable region sequences set forth in SEQ ID NOS 16 and 18, respectivelyLight chain variable region sequences. The heavy chain variable regions of mAbs PDL1#1 and PDL1#10 are encoded by the nucleic acid sequences set forth in SEQ ID NOS 13 and 15, respectively, and the light chain variable regions of mAbs PDL1#1 and PDL1#10 are encoded by the nucleic acid sequences set forth in SEQ ID NOS 17 and 19, respectively.
Pdl1#10 was evaluated in a Mixed Lymphocyte Reaction (MLR) assay to evaluate the efficacy of pdl1#10 on T cell activation. T cell activation is measured by the concentration of interleukin-2 (IL-2) secreted by T cells. Isolation of Dendritic Cells (DC) and CD 4 from human Peripheral Blood Mononuclear Cells (PBMC) of healthy donors + T cells. CD4 + T cell passage through CD4 + Purifying the T cell separation kit. Dendritic cells were purified with Pan monocyte isolation kit and analyzed in FACS assays to determine their co-stimulatory molecules and MHC class II expression. It was determined that DC was stimulated and the expression of its surface markers CD1a, CD83, CD86 and H LA-DR was observed. In MLR(Merck&Co.) efficacy on T cell activation was used as an internal control to monitor assay performance. Half maximum effective concentration (EC 50 ) Values were analyzed by GraphPad Prism, fitted with Sigmoidal dose-response nonlinear regression. EC of PDL1#10 on T cell activation 50 The value was.104. Mu.g/mL, whereas the internal control was +.>EC on T cell activation 50 The value was.19. Mu.g/mL. IC for T cell activation by PDL1#10 50 The value is 0.2nM, whereas the internal control +.>IC for T cell activation 50 The value was 1.5nM. This demonstrates that pdl1#10 is able to increase T cell proliferation and IL-2 secretion in mixed lymphocyte responses.
A murine-human chimeric Fab (hereinafter "chimeric Fab") was prepared using the HCVR sequence and LCVR sequence of mAb PDL1#10, and comprises the heavy chain sequence set forth in SEQ ID NO:51 and the light chain sequence set forth in SEQ ID NO: 53. The heavy and light chains of the chimeric Fab are encoded by the nucleic acids set forth in SEQ ID NOS 52 and 54, respectively.
Example 2
Production of humanized Fab specifically targeting human PD-L1
The combined humanized phage display library was constructed using the heavy chain variable domain sequence and the light chain variable domain sequence of mAb pdl1#10. Briefly, a framework of human antibodies with high sequence identity to pdl1#10 was selected and assembled using overlap PCR. The humanized Fab phage display library was panned (panned) against the PD-L1 Fc fusion protein. Individual output phage clones were amplified in 96-deep well plates and the amplified phage was assayed by ELISA against PD-L1 Fc fusion proteins. Panning was ended when a good percentage of phage clones were found to bind to PD-L1 protein. DNA encoding Fab fragments of output phage was amplified and inserted into pFASEBA vector (Genscript) for selection of optimally humanized antibody clones. Individual Fab clones were expressed in 96 deep-well plates and crude proteins secreted by e.coli into the medium were assayed by ELISA against BSA and the antigen PD-L1 protein for evaluation of expression and binding activity, respectively. Thousands of humanized clones were screened and many clones with lower dissociation rates (off-rates) were identified, with the 14 clones with the highest binding affinity undergoing multicycle affinity measurements. Binding affinity of 14 purified fabs to His-tagged PD-L1 proteins was determined using the Surface Plasmon Resonance (SPR) biosensor Biacore T200 (GE Healthcare) for multicycle affinity measurement. Fab was immobilized on the sensor chip by capture. His-tagged antigen PD-L1 protein was used as analyte. Data for the dissociation (kd) and association (ka) rate constants were understood at the evaluation software using Biacore T200. The equilibrium dissociation constant (KD) was calculated from the KD to ka ratio. The results are depicted in table 4 below:
TABLE 4 Table 4
Fab Ka(1/Ms) Kd(1/s) KD(M)
AS01790 5.0E+05 1.7E-04 3.5E-10
AS02013 5.0E+05 2.0E-04 3.8E-10
AS01981 5.2E+05 1.5E-04 2.8E-10
AS01798 5.5E+05 1.9E-04 3.5E-10
AS01814 6.8E+05 1.1E-04 1.6E-10
AS01789 3.3E+05 3.6E-04 1.1E-09
AS01995 4.5E+05 1.7E-04 3.7E-10
AS01979 4.7E+05 1.9E-04 4.0E-10
AS01817 3.4E+05 3.2E-04 9.3E-10
AS01824 6.2E+05 5.4E-04 8.7E-10
AS01807 2.6E+05 3.5E-04 1.4E-09
AS01825 3.6E+05 3.9E-04 1.1E-09
AS02002 3.1E+05 4.2E-04 1.5E-09
AS01978 2.9E+05 4.5E-04 1.6E-09
Chimeric Fab 7.3E+05 2.1E-04 2.9E-10
HCVR and LCVR for 14 humanized Fab are depicted in table 5:
TABLE 5
Fab HCVR LCVR
AS01790 SEQ ID NO:20 SEQ ID NO:21
AS02013 SEQ ID NO:22 SEQ ID NO:23
AS01981 SEQ ID NO:24 SEQ ID NO:25
AS01798 SEQ ID NO:26 SEQ ID NO:27
AS01814 SEQ ID NO:28 SEQ ID NO:29
AS01789 SEQ ID NO:30 SEQ ID NO:31
AS01995 SEQ ID NO:32 SEQ ID NO:33
AS01979 SEQ ID NO:34 SEQ ID NO:35
AS01817 SEQ ID NO:36 SEQ ID NO:37
AS01824 SEQ ID NO:38 SEQ ID NO:39
AS01807 SEQ ID NO:40 SEQ ID NO:41
AS01825 SEQ ID NO:42 SEQ ID NO:43
AS02002 SEQ ID NO:44 SEQ ID NO:45
AS01978 SEQ ID NO:46 SEQ ID NO:47
Example 3
Production of humanized IgG specifically targeting human PD-L1
Based on the results of the affinity ranking and multicycle affinity ranking measurements, AS01814, AS01981, AS02013, AS01790 and AS01798 clones were selected for full length IgG expression, purification and affinity measurement. DNA sequences encoding AS01814, AS01981, AS02013, AS01790 and AS01798 (including leader sequences) were amplified and inserted into pTT5 to prepare expression plasmids for full-length IgG. The heavy and light chain expression plasmids were used to co-transfect HEK293 cells (100 ml cell culture). Recombinant IgG secreted into the medium was purified using protein a affinity chromatography. Purified IgG migrates as a band of 170kDa in SDS-PAGE under non-reducing conditions, and as a band of 55kDa and a band of 30kDa in SDS-PAGE under reducing conditions. The purity, yield and endotoxin levels of each humanized clone are depicted in table 6 below.
TABLE 6
/>
Binding data for each antibody was processed using Biacore T200 evaluation software and fitted to a 1:1 interaction model. All experimental data can be well fitted to the model. AS can be seen from table 7, all 5 humanized antibodies (AS 01790, AS01798, AS01814, AS01981 and AS 02013) have higher antigen binding affinity than mouse-human chimeric antibodies.
TABLE 7
IgG Ka(1/Ms) Kd(1/s) KD(M)
AS01790 3.29E+05 2.53E-04 7.69E-10
AS01798 3.58E+05 2.72E-04 7.59E-10
AS01814 3.76E+05 1.73E-04 4.61E-10
AS01981 3.05E+05 1.99E-04 6.53E-10
AS02013 3.67E+05 2.76E-04 7.51E-10
Chimeric IgG 3.36E+05 3.38E-04 1.00E-09
The amino acid sequences of HC and LC of 5 humanized IgG are depicted in table 8:
TABLE 8
Fab HC LC
AS01790 SEQ ID NO:55 SEQ ID NO:56
AS02013 SEQ ID NO:57 SEQ ID NO:58
AS01981 SEQ ID NO:59 SEQ ID NO:60
AS01798 SEQ ID NO:61 SEQ ID NO:62
AS01814 SEQ ID NO:63 SEQ ID NO:64
Example 4
Evaluation of PD-L1 mAb efficacy by Mixed lymphocyte reaction
A Mixed Lymphocyte Reaction (MLR) assay was performed to evaluate the efficacy of chimeric Fab, AS01790, AS01814 and AS01981 on T cell activation. Activation of T cells is measured by the concentration of interleukin 2 (IL-2) secreted by T cells. Dendritic Cells (DCs) and CD 4 + T cells were isolated from human Peripheral Blood Mononuclear Cells (PBMCs). In MLREfficacy on T cell activation was used as an internal control to monitor assay performance. Half maximal Effective Concentration (EC) was analyzed by GraphPad Prism, fitting with agonist dose-response variable slope (four parameters) 50 ) Values. EC of chimeric Fab, AS01790, AS01814 and AS01981 on T cell activation 50 The values were 1.048. Mu.g/ml, 0.174. Mu.g/ml, 0.089. Mu.g/ml and 0.106. Mu.g/ml, respectively. Negative control, human IgG4, did not have any effect on T cell activation. T cell activation data from the internal control (Keytruda activation of T cells in MLR) was consistent with historical data of GenSript (.19 μg/ml) in all five plates, thus confirming the study. This demonstrates that humanized PD-L1 antibodies are able to increase T cell proliferation and IL-2 secretion in mixed lymphocyte responses.
All of the articles and methods disclosed and claimed herein can be made and executed without undue experimentation in light of the present disclosure. While the article and method of the present invention have been described in terms of preferred embodiments, it will be apparent to those of skill in the art that variations may be applied to the article and method without departing from the spirit and scope of the invention. All such variations and equivalents that are apparent to a person skilled in the art, whether existing or later to be developed, are considered to be within the spirit and scope of the present invention as defined by the appended claims. All patents, patent applications, and publications mentioned in the specification are indicative of the levels of those of ordinary skill in the art to which the invention pertains. All patents, patent applications, and publications are herein incorporated by reference in their entirety for all purposes to the same extent as if each individual publication was specifically and individually indicated to be incorporated by reference in its entirety for any and all purposes. The invention illustratively described herein suitably may be practiced in the absence of any element or elements not specifically disclosed herein. Therefore, it should be understood that although the present invention has been specifically disclosed by preferred embodiments and optional features, modification and variation of the concepts herein disclosed may be resorted to by those skilled in the art, and that such modifications and variations are considered to be within the scope of this invention as defined by the appended claims.
Sequence listing
The nucleic acid and amino acid sequences listed in the accompanying sequence listing are shown using standard alphabetical abbreviations for nucleotide bases and three letter coding for amino acids as specified in 37 c.f.r.1.822.
SEQ ID NO. 1 is the amino acid sequence of a human PD-L1 polypeptide.
SEQ ID NO. 2 is the amino acid sequence of the heavy chain CDR1 in a monoclonal antibody that specifically binds PD-L1.
SEQ ID NO. 3-4 is the amino acid sequence of the heavy chain CDR2 in a monoclonal antibody that specifically binds PD-L1.
SEQ ID NO. 5-6 is the amino acid sequence of the heavy chain CDR3 of a monoclonal antibody that specifically binds PD-L1.
SEQ ID NO. 7-8 are amino acid sequences of the light chain CDR1 in a monoclonal antibody that specifically binds PD-L1.
SEQ ID NO. 9 is the amino acid sequence of the light chain CDR2 of a monoclonal antibody that specifically binds PD-L1.
SEQ ID NO. 10-11 is the amino acid sequence of the light chain CDR3 of a monoclonal antibody that specifically binds PD-L1.
SEQ ID NOS 12 and 14 are amino acid sequences of the heavy chain variable region of a murine monoclonal antibody that specifically binds to PD-L1.
SEQ ID NOS 13 and 15 are nucleic acid sequences encoding the heavy chain variable region of a murine monoclonal antibody that specifically binds to PD-L1.
SEQ ID NOS.16 and 18 are amino acid sequences of the light chain variable region of a murine monoclonal antibody that specifically binds to PD-L1.
SEQ ID NOS.17 and 19 are nucleic acid sequences encoding the light chain variable region of a murine monoclonal antibody that specifically binds to PD-L1.
20, 22, 24, 26, 28, 30, 32, 34, 36, 38, 40, 42, 44 and 46 are the amino acid sequences of the heavy chain variable region of a humanized monoclonal antibody that specifically binds to PD-L1.
21, 23, 25, 27, 29, 31, 33, 35, 37, 39, 41, 43, 45 and 47 are the amino acid sequences of the light chain variable region of a humanized monoclonal antibody that specifically binds to PD-L1.
SEQ ID NOS.48 and 49 are amino acid sequences of the light chain constant region of the monoclonal antibody.
SEQ ID NO. 50 is the amino acid sequence of the heavy chain constant region of the monoclonal antibody.
SEQ ID NO. 51 is the amino acid sequence of the heavy chain of a murine-human chimeric antibody that specifically binds to PD-L1.
SEQ ID NO. 52 is a nucleic acid sequence of the heavy chain of a murine-human chimeric antibody that specifically binds to PD-L1.
SEQ ID NO. 53 is the amino acid sequence of the light chain of the murine-human chimeric antibody that specifically binds to PD-L1.
SEQ ID NO. 54 is a nucleic acid sequence of the light chain of a murine-human chimeric antibody that specifically binds to PD-L1.
SEQ ID NOS: 55, 57, 59, 61 and 63 are amino acid sequences of heavy chains of humanized monoclonal antibodies that specifically bind to PD-L1.
SEQ ID NOS 56, 58, 60, 62 and 64 are amino acid sequences of the light chain of a humanized monoclonal antibody that specifically binds to PD-L1.
SEQ ID NOS.65, 67, 69, 71 and 73 are nucleic acid sequences of the heavy chains of humanized monoclonal antibodies that specifically bind to PD-L1.
SEQ ID NOS.66, 68, 70, 72 and 74 are nucleic acid sequences of the light chain of a humanized monoclonal antibody that specifically binds to PD-L1.
Sequence listing
SEQ ID NO. 1-PD-L1 antigen amino acid sequence
SEQ ID NO. 2-murine monoclonal antibody heavy chain CDR1 amino acid sequence
SYSIN
SEQ ID NO. 3-murine monoclonal antibody heavy chain CDR2 amino acid sequence
YIYIGNGYTEYNEKFKG
SEQ ID NO. 4-murine monoclonal antibody heavy chain CDR2 amino acid sequence
YFYVGNGYTDYNEKFKG
SEQ ID NO. 5-murine monoclonal antibody heavy chain CDR3 amino acid sequence
WSLPNGMDY
SEQ ID NO. 6-murine monoclonal antibody heavy chain CDR3 amino acid sequence
GGLPYYFDY
SEQ ID NO. 7-murine monoclonal antibody light chain CDR1 amino acid sequence
KASQDVGISVA
SEQ ID NO 8-murine monoclonal antibody light chain CDR1 amino acid sequence
KTSQDVNTAVA
SEQ ID NO 9-murine monoclonal antibody light chain CDR2 amino acid sequence
WASTRHT
SEQ ID NO. 10-murine monoclonal antibody light chain CDR3 amino acid sequence
QQYSTYRT
11-murine monoclonal antibody light chain CDR3 amino acid sequence
QQHYNTPLT
SEQ ID NO. 12-mouse monoclonal antibody heavy chain variable region amino acid sequence
SEQ ID NO. 13-murine monoclonal heavy chain variable region nucleic acid sequence
SEQ ID NO. 14-murine monoclonal antibody heavy chain variable region amino acid sequence
15-murine monoclonal heavy chain variable region nucleic acid sequence
SEQ ID NO. 16-murine monoclonal antibody light chain variable region amino acid sequence
SEQ ID NO. 17-murine monoclonal light chain variable region nucleic acid sequence
SEQ ID NO. 18-murine monoclonal antibody light chain variable region amino acid sequence
SEQ ID NO. 19-murine monoclonal light chain variable region nucleic acid sequence
SEQ ID NO. 20-humanized heavy chain variable region amino acid sequence
SEQ ID NO. 21-humanized light chain variable region amino acid sequence
SEQ ID NO. 22-humanized heavy chain variable region amino acid sequence
SEQ ID NO. 23-humanized light chain variable region amino acid sequence
SEQ ID NO. 24-humanized heavy chain variable region amino acid sequence
25-humanized light chain variable region amino acid sequence of SEQ ID NO
SEQ ID NO. 26-humanized heavy chain variable region amino acid sequence
SEQ ID NO. 27-humanized light chain variable region amino acid sequence
28-humanized heavy chain variable region amino acid sequence of SEQ ID NO
SEQ ID NO. 29-humanized light chain variable region amino acid sequence
SEQ ID NO. 30-humanized heavy chain variable region amino acid sequence
SEQ ID NO. 31-humanized light chain variable region amino acid sequence
SEQ ID NO. 32-humanized heavy chain variable region amino acid sequence
33-humanized light chain variable region amino acid sequence of SEQ ID NO
SEQ ID NO. 34-humanized heavy chain variable region amino acid sequence
SEQ ID NO. 35-humanized light chain variable region amino acid sequence
SEQ ID NO. 36-humanized heavy chain variable region amino acid sequence
SEQ ID NO. 37-humanized light chain variable region amino acid sequence
38-humanized heavy chain variable region amino acid sequence of SEQ ID NO
SEQ ID NO. 39-humanized light chain variable region amino acid sequence
SEQ ID NO. 40-humanized heavy chain variable region amino acid sequence
SEQ ID NO. 41-humanized light chain variable region amino acid sequence
/>
SEQ ID NO. 42-humanized heavy chain variable region amino acid sequence
43-humanized light chain variable region amino acid sequence of SEQ ID NO
SEQ ID NO. 44-humanized heavy chain variable region amino acid sequence
SEQ ID NO. 45-humanized light chain variable region amino acid sequence
46-humanized heavy chain variable region amino acid sequence of SEQ ID NO
SEQ ID NO. 47-humanized light chain variable region amino acid sequence
SEQ ID NO. 48-constant region amino acid sequence of light chain
49-light chain constant region amino acid sequence of SEQ ID NO
SEQ ID NO. 50-heavy chain constant region amino acid sequence
Heavy chain amino acid sequence of SEQ ID NO. 51-mouse-human chimeric antibody
Heavy chain nucleic acid sequence of 52-mouse-human chimeric antibody
Light chain amino acid sequence of the 53-mouse-human chimeric antibody
Light chain nucleic acid sequence of 54-mouse-human chimeric antibody
55-humanized heavy chain amino acid sequence of SEQ ID NO
SEQ ID NO. 56-humanized light chain amino acid sequence
SEQ ID NO. 57-humanized heavy chain amino acid sequence
/>
SEQ ID NO. 58-humanized light chain amino acid sequence
59-humanized heavy chain amino acid sequence of SEQ ID NO
SEQ ID NO. 60-humanized light chain amino acid sequence
SEQ ID NO. 61-humanized heavy chain amino acid sequence
SEQ ID NO. 62-humanized light chain amino acid sequence
63-humanized heavy chain amino acid sequence of SEQ ID NO
SEQ ID NO. 64-humanized light chain amino acid sequence
SEQ ID NO. 65-humanized heavy chain nucleic acid sequence
SEQ ID NO. 66-humanized light chain nucleic acid sequence
SEQ ID NO. 67-humanized heavy chain nucleic acid sequence
68-humanized light chain nucleic acid sequence of SEQ ID NO
69-humanized heavy chain nucleic acid sequence
70-humanized light chain nucleic acid sequence of SEQ ID NO
SEQ ID NO. 71-humanized heavy chain nucleic acid sequence
SEQ ID NO. 72-humanized light chain nucleic acid sequence
73-humanized heavy chain nucleic acid sequence of SEQ ID NO
74-humanized light chain nucleic acid sequence of SEQ ID NO
/>
Sequence listing
<110> Ruimedes biological medicine technology Co., ltd
<120> immunotherapy using antibodies that bind to programmed death ligand 1 (PD-L1)
<130> CACRE1.0012WO
<160> 74
<170> patent In version 3.5
<210> 1
<211> 290
<212> PRT
<213> Homo sapiens (Homo sapiens)
<400> 1
Met Arg Ile Phe Ala Val Phe Ile Phe Met Thr Tyr Trp His Leu Leu
1 5 10 15
Asn Ala Phe Thr Val Thr Val Pro Lys Asp Leu Tyr Val Val Glu Tyr
20 25 30
Gly Ser Asn Met Thr Ile Glu Cys Lys Phe Pro Val Glu Lys Gln Leu
35 40 45
Asp Leu Ala Ala Leu Ile Val Tyr Trp Glu Met Glu Asp Lys Asn Ile
50 55 60
Ile Gln Phe Val His Gly Glu Glu Asp Leu Lys Val Gln His Ser Ser
65 70 75 80
Tyr Arg Gln Arg Ala Arg Leu Leu Lys Asp Gln Leu Ser Leu Gly Asn
85 90 95
Ala Ala Leu Gln Ile Thr Asp Val Lys Leu Gln Asp Ala Gly Val Tyr
100 105 110
Arg Cys Met Ile Ser Tyr Gly Gly Ala Asp Tyr Lys Arg Ile Thr Val
115 120 125
Lys Val Asn Ala Pro Tyr Asn Lys Ile Asn Gln Arg Ile Leu Val Val
130 135 140
Asp Pro Val Thr Ser Glu His Glu Leu Thr Cys Gln Ala Glu Gly Tyr
145 150 155 160
Pro Lys Ala Glu Val Ile Trp Thr Ser Ser Asp His Gln Val Leu Ser
165 170 175
Gly Lys Thr Thr Thr Thr Asn Ser Lys Arg Glu Glu Lys Leu Phe Asn
180 185 190
Val Thr Ser Thr Leu Arg Ile Asn Thr Thr Thr Asn Glu Ile Phe Tyr
195 200 205
Cys Thr Phe Arg Arg Leu Asp Pro Glu Glu Asn His Thr Ala Glu Leu
210 215 220
Val Ile Pro Glu Leu Pro Leu Ala His Pro Pro Asn Glu Arg Thr His
225 230 235 240
Leu Val Ile Leu Gly Ala Ile Leu Leu Cys Leu Gly Val Ala Leu Thr
245 250 255
Phe Ile Phe Arg Leu Arg Lys Gly Arg Met Met Asp Val Lys Lys Cys
260 265 270
Gly Ile Gln Asp Thr Asn Ser Lys Lys Gln Ser Asp Thr His Leu Glu
275 280 285
Glu Thr
290
<210> 2
<211> 5
<212> PRT
<213> mice (Mus musculus)
<400> 2
Ser Tyr Ser Ile Asn
1 5
<210> 3
<211> 17
<212> PRT
<213> mice (Mus musculus)
<400> 3
Tyr Ile Tyr Ile Gly Asn Gly Tyr Thr Glu Tyr Asn Glu Lys Phe Lys
1 5 10 15
Gly
<210> 4
<211> 17
<212> PRT
<213> mice (Mus musculus)
<400> 4
Tyr Phe Tyr Val Gly Asn Gly Tyr Thr Asp Tyr Asn Glu Lys Phe Lys
1 5 10 15
Gly
<210> 5
<211> 9
<212> PRT
<213> mice (Mus musculus)
<400> 5
Trp Ser Leu Pro Asn Gly Met Asp Tyr
1 5
<210> 6
<211> 9
<212> PRT
<213> mice (Mus musculus)
<400> 6
Gly Gly Leu Pro Tyr Tyr Phe Asp Tyr
1 5
<210> 7
<211> 11
<212> PRT
<213> mice (Mus musculus)
<400> 7
Lys Ala Ser Gln Asp Val Gly Ile Ser Val Ala
1 5 10
<210> 8
<211> 11
<212> PRT
<213> mice (Mus musculus)
<400> 8
Lys Thr Ser Gln Asp Val Asn Thr Ala Val Ala
1 5 10
<210> 9
<211> 7
<212> PRT
<213> mice (Mus musculus)
<400> 9
Trp Ala Ser Thr Arg His Thr
1 5
<210> 10
<211> 8
<212> PRT
<213> mice (Mus musculus)
<400> 10
Gln Gln Tyr Ser Thr Tyr Arg Thr
1 5
<210> 11
<211> 9
<212> PRT
<213> mice (Mus musculus)
<400> 11
Gln Gln His Tyr Asn Thr Pro Leu Thr
1 5
<210> 12
<211> 118
<212> PRT
<213> mice (Mus musculus)
<400> 12
Glu Val Gln Leu Gln Gln Ser Gly Ala Glu Leu Val Arg Pro Gly Ser
1 5 10 15
Ser Val Lys Met Ser Cys Lys Thr Ser Gly Tyr Ser Phe Thr Ser Tyr
20 25 30
Ser Ile Asn Trp Val Lys Gln Arg Pro Gly Gln Gly Leu Glu Trp Val
35 40 45
Ala Tyr Ile Tyr Ile Gly Asn Gly Tyr Thr Glu Tyr Asn Glu Lys Phe
50 55 60
Lys Gly Lys Ala Thr Leu Thr Ser Asp Thr Pro Ser Ser Thr Ala Tyr
65 70 75 80
Met Gln Leu Ser Ser Leu Thr Ser Glu Asp Ser Ala Ile Tyr Phe Cys
85 90 95
Ala Arg Trp Ser Leu Pro Asn Gly Met Asp Tyr Trp Gly Gln Gly Thr
100 105 110
Ser Val Thr Val Ser Ser
115
<210> 13
<211> 354
<212> DNA
<213> mice (Mus musculus)
<400> 13
gaggtccagc ttcagcagtc tggagctgaa ctggtgaggc ctgggtcctc agtgaagatg 60
tcctgcaaga cttctggata ttcattcaca agttacagta taaactgggt gaagcagagg 120
cctggacagg gcctggaatg ggttgcatat atttatattg gaaatggtta tactgaatat 180
aatgagaagt tcaagggcaa ggccacactg acttcagaca caccctccag cacagcctac 240
atgcagctca gcagcctgac atctgaggat tctgcaatct atttctgtgc aagatggtca 300
ctacccaatg gaatggacta ctgggggcaa ggaacctcag tcaccgtctc ctca 354
<210> 14
<211> 118
<212> PRT
<213> mice (Mus musculus)
<400> 14
Glu Val Gln Leu Gln Gln Ser Gly Ala Glu Leu Val Arg Pro Gly Ser
1 5 10 15
Ser Val Lys Met Ser Cys Lys Thr Ser Gly Tyr Thr Phe Thr Ser Tyr
20 25 30
Ser Ile Asn Trp Val Lys Gln Arg Pro Gly Gln Gly Leu Glu Trp Ile
35 40 45
Ala Tyr Phe Tyr Val Gly Asn Gly Tyr Thr Asp Tyr Asn Glu Lys Phe
50 55 60
Lys Gly Lys Ala Thr Leu Thr Ser Asp Thr Ser Ser Ser Thr Ala Tyr
65 70 75 80
Met Gln Leu Ser Ser Leu Thr Ser Glu Asp Ser Ala Ile Tyr Phe Cys
85 90 95
Ala Arg Gly Gly Leu Pro Tyr Tyr Phe Asp Tyr Trp Gly Gln Gly Thr
100 105 110
Thr Leu Thr Val Ser Ser
115
<210> 15
<211> 354
<212> DNA
<213> mice (Mus musculus)
<400> 15
gaggtccagc ttcagcagtc tggagctgaa ctggtgaggc ctgggtcctc agtgaagatg 60
tcctgcaaga cttctggata tacattcaca agttacagta taaattgggt gaagcagagg 120
cctggtcagg gcctggaatg gattgcatat ttttatgttg gaaatggtta tactgactac 180
aatgagaagt tcaagggcaa ggccacactg acttcagaca catcttccag cacagcctac 240
atgcagctca gcagcctgac atctgaggac tctgcaatct atttctgtgc acgagggggc 300
ctaccctact actttgacta ctggggccaa ggcaccactc tcacagtctc ctca 354
<210> 16
<211> 106
<212> PRT
<213> mice (Mus musculus)
<400> 16
Asp Ile Val Met Thr Gln Ser His Lys Leu Met Ser Thr Ser Ile Gly
1 5 10 15
Asp Arg Val Asn Ile Thr Cys Lys Ala Ser Gln Asp Val Gly Ile Ser
20 25 30
Val Ala Trp Tyr Gln Gln Lys Pro Gly Gln Ser Pro Lys Leu Leu Ile
35 40 45
Tyr Trp Ala Ser Thr Arg His Thr Gly Val Pro Asp Arg Phe Thr Gly
50 55 60
Ser Gly Ser Gly Thr Asp Phe Thr Leu Thr Ile Ser Asn Val Gln Ser
65 70 75 80
Glu Asp Leu Ala Asp Tyr Phe Cys Gln Gln Tyr Ser Thr Tyr Arg Thr
85 90 95
Phe Gly Gly Gly Thr Lys Leu Glu Asn Lys
100 105
<210> 17
<211> 318
<212> DNA
<213> mice (Mus musculus)
<400> 17
gacattgtga tgacccagtc tcacaaactc atgtccacat caataggaga cagggtcaac 60
atcacctgca aggccagtca ggatgtgggt atttctgtag cctggtatca acagaaacca 120
ggacaatctc ctaaactact gatttattgg gcatccactc ggcacactgg agtccccgat 180
cgcttcacag gcagtggatc tgggacagat ttcactctca ccattagtaa tgtgcagtct 240
gaggacttgg cagattattt ctgtcagcaa tatagcacct atcggacgtt cggtggaggc 300
accaagctgg aaaacaaa 318
<210> 18
<211> 107
<212> PRT
<213> mice (Mus musculus)
<400> 18
Asp Ile Val Met Thr Gln Ser His Lys Phe Met Ser Thr Ser Val Gly
1 5 10 15
Asp Arg Val Ser Ile Thr Cys Lys Thr Ser Gln Asp Val Asn Thr Ala
20 25 30
Val Ala Trp Tyr Gln Gln Lys Pro Gly His Ser Pro Lys Leu Leu Phe
35 40 45
Tyr Trp Ala Ser Thr Arg His Thr Gly Val Pro Asp Arg Phe Thr Gly
50 55 60
Gly Gly Ser Gly Thr Asp Tyr Thr Leu Thr Ile Ser Ser Val Gln Ala
65 70 75 80
Glu Asp Leu Ala Leu Tyr Tyr Cys Gln Gln His Tyr Asn Thr Pro Leu
85 90 95
Thr Phe Gly Ala Gly Thr Arg Leu Glu Leu Lys
100 105
<210> 19
<211> 321
<212> DNA
<213> mice (Mus musculus)
<400> 19
gacattgtga tgacccagtc tcacaaattc atgtccacat cagttggaga cagggtcagc 60
atcacctgca agaccagtca ggatgtgaat actgctgtag cctggtatca acaaaaacca 120
gggcattctc ctaaactact gttttactgg gcatccaccc ggcacactgg agtccctgat 180
cgcttcacag gcggtggatc tgggacagat tatactctca ccatcagcag tgtgcaggct 240
gaagacctgg cactttatta ctgtcagcaa cattataaca ctccgctcac gttcggtgct 300
gggaccaggc tggagctgaa a 321
<210> 20
<211> 118
<212> PRT
<213> Artificial (Artifical)
<220>
<223> heavy chain variable region of humanized antibody binding to PD-L1
<400> 20
Glu Val Gln Leu Val Gln Ser Gly Ala Glu Val Lys Lys Pro Gly Glu
1 5 10 15
Ser Leu Lys Ile Ser Cys Lys Gly Ser Gly Tyr Ser Phe Thr Ser Tyr
20 25 30
Ser Ile Asn Trp Val Arg Gln Met Pro Gly Lys Gly Leu Glu Trp Met
35 40 45
Gly Tyr Phe Tyr Val Gly Asn Gly Tyr Thr Asp Tyr Asn Glu Lys Phe
50 55 60
Lys Gly Arg Val Thr Met Thr Arg Asn Thr Ser Ile Ser Thr Ala Tyr
65 70 75 80
Met Glu Leu Ser Ser Leu Arg Ser Glu Asp Thr Ala Val Tyr Tyr Cys
85 90 95
Ala Arg Gly Gly Leu Pro Tyr Tyr Phe Asp Tyr Trp Gly Gln Gly Thr
100 105 110
Leu Val Thr Val Ser Ser
115
<210> 21
<211> 107
<212> PRT
<213> Artificial (Artifical)
<220>
<223> light chain variable region of humanized antibody binding to PD-L1
<400> 21
Asp Ile Gln Met Thr Gln Ser Pro Ser Thr Leu Ser Ala Ser Val Gly
1 5 10 15
Asp Arg Val Thr Ile Thr Cys Lys Thr Ser Gln Asp Val Asn Thr Ala
20 25 30
Val Ala Trp Tyr Gln Gln Lys Pro Gly Lys Ala Pro Lys Leu Leu Ile
35 40 45
Tyr Trp Ala Ser Thr Arg His Thr Gly Val Pro Ser Arg Phe Ser Gly
50 55 60
Ser Gly Ser Gly Thr Glu Phe Thr Leu Thr Ile Ser Ser Leu Gln Pro
65 70 75 80
Glu Asp Phe Ala Thr Tyr Tyr Cys Gln Gln His Tyr Asn Thr Pro Leu
85 90 95
Thr Phe Gly Gly Gly Thr Lys Leu Glu Ile Lys
100 105
<210> 22
<211> 117
<212> PRT
<213> Artificial (Artifical)
<220>
<223> heavy chain variable region of humanized antibody binding to PD-L1
<400> 22
Glu Val Gln Leu Val Gln Ser Gly Ala Glu Val Lys Lys Pro Gly Glu
1 5 10 15
Ser Leu Lys Ile Ser Cys Lys Gly Ser Gly Tyr Ser Phe Thr Ser Tyr
20 25 30
Ser Ile Asn Trp Val Arg Gln Ala Pro Gly Lys Gly Leu Glu Trp Met
35 40 45
Gly Tyr Phe Tyr Val Gly Asn Gly Tyr Thr Asp Tyr Asn Glu Lys Phe
50 55 60
Lys Gly Arg Val Thr Met Thr Arg Asn Thr Ser Ile Ser Thr Ala Tyr
65 70 75 80
Met Glu Leu Ser Ser Leu Arg Ser Glu Asp Thr Ala Val Tyr Tyr Cys
85 90 95
Ala Arg Gly Gly Leu Pro Tyr Tyr Phe Asp Tyr Gly Gln Gly Thr Leu
100 105 110
Val Thr Val Ser Ser
115
<210> 23
<211> 107
<212> PRT
<213> Artificial (Artifical)
<220>
<223> light chain variable region of humanized antibody binding to PD-L1
<400> 23
Asp Ile Gln Leu Thr Gln Ser Pro Ser Ser Leu Ser Ala Ser Val Gly
1 5 10 15
Asp Arg Val Thr Ile Thr Cys Lys Thr Ser Gln Asp Val Asn Thr Ala
20 25 30
Val Ala Trp Tyr Gln Gln Lys Pro Gly Gln Ala Pro Arg Leu Leu Ile
35 40 45
Tyr Trp Ala Ser Thr Arg His Thr Gly Val Pro Ser Arg Phe Ser Gly
50 55 60
Ser Gly Ser Gly Thr Asp Phe Thr Leu Thr Ile Ser Ser Leu Gln Pro
65 70 75 80
Glu Asp Phe Ala Thr Tyr Tyr Cys Gln Gln His Tyr Asn Thr Pro Leu
85 90 95
Thr Phe Gly Gly Gly Thr Lys Leu Glu Ile Lys
100 105
<210> 24
<211> 118
<212> PRT
<213> Artificial (Artifical)
<220>
<223> heavy chain variable region of humanized antibody binding to PD-L1
<400> 24
Gln Met Gln Leu Val Gln Ser Gly Ala Glu Val Lys Lys Thr Gly Ser
1 5 10 15
Ser Val Lys Val Ser Cys Lys Ala Ser Gly Tyr Thr Phe Thr Ser Tyr
20 25 30
Ser Ile Asn Trp Val Arg Gln Ala Pro Gly Lys Gly Leu Glu Trp Val
35 40 45
Ala Tyr Phe Tyr Val Gly Asn Gly Tyr Thr Asp Tyr Asn Glu Lys Phe
50 55 60
Lys Gly Arg Val Thr Met Thr Arg Asp Thr Ser Thr Ser Thr Val Tyr
65 70 75 80
Met Glu Leu Ser Ser Leu Arg Ser Glu Asp Thr Ala Val Tyr Tyr Cys
85 90 95
Ala Arg Gly Gly Leu Pro Tyr Tyr Phe Asp Tyr Trp Gly Gln Gly Thr
100 105 110
Leu Val Thr Val Ser Ser
115
<210> 25
<211> 107
<212> PRT
<213> Artificial (Artifical)
<220>
<223> light chain variable region of humanized antibody binding to PD-L1
<400> 25
Asp Ile Gln Met Thr Gln Ser Pro Ser Ser Leu Ser Ala Ser Val Gly
1 5 10 15
Asp Arg Val Thr Ile Thr Cys Lys Thr Ser Gln Asp Val Asn Thr Ala
20 25 30
Val Ala Trp Tyr Gln Gln Lys Pro Gly Gln Pro Pro Lys Leu Leu Ile
35 40 45
Tyr Trp Ala Ser Thr Arg His Thr Gly Ile Pro Ala Arg Phe Ser Gly
50 55 60
Ser Gly Ser Gly Thr Asp Phe Thr Leu Thr Ile Ser Ser Leu Gln Pro
65 70 75 80
Glu Asp Phe Ala Val Tyr Tyr Cys Gln Gln His Tyr Asn Thr Pro Leu
85 90 95
Thr Phe Gly Gly Gly Thr Lys Leu Glu Ile Lys
100 105
<210> 26
<211> 118
<212> PRT
<213> Artificial (Artifical)
<220>
<223> heavy chain variable region of humanized antibody binding to PD-L1
<400> 26
Gln Val Gln Leu Val Gln Ser Gly Ala Glu Val Lys Lys Pro Gly Ala
1 5 10 15
Ser Val Lys Val Ser Cys Lys Ala Ser Gly Tyr Thr Phe Thr Ser Tyr
20 25 30
Ser Ile Asn Trp Val Arg Gln Ala Pro Gly Lys Gly Leu Glu Trp Val
35 40 45
Ala Tyr Phe Tyr Val Gly Asn Gly Tyr Thr Asp Tyr Asn Glu Lys Phe
50 55 60
Lys Gly Arg Val Thr Ile Thr Thr Asp Glu Ser Thr Ser Thr Ala Tyr
65 70 75 80
Met Glu Leu Ser Ser Leu Arg Ser Glu Asp Thr Ala Val Tyr Tyr Cys
85 90 95
Ala Arg Gly Gly Leu Pro Tyr Tyr Phe Asp Tyr Trp Gly Gln Gly Thr
100 105 110
Leu Val Thr Val Ser Ser
115
<210> 27
<211> 107
<212> PRT
<213> Artificial (Artifical)
<220>
<223> light chain variable region of humanized antibody binding to PD-L1
<400> 27
Asp Ile Gln Met Thr Gln Ser Pro Ser Ser Val Ser Ala Ser Val Gly
1 5 10 15
Asp Arg Val Thr Ile Thr Cys Lys Thr Ser Gln Asp Val Asn Thr Ala
20 25 30
Val Ala Trp Tyr Gln Gln Lys Pro Gly Gln Ala Pro Arg Leu Leu Ile
35 40 45
Tyr Trp Ala Ser Thr Arg His Thr Gly Val Pro Asp Arg Phe Ser Gly
50 55 60
Ser Gly Ser Gly Thr Asp Phe Thr Leu Thr Ile Ser Ser Leu Gln Ala
65 70 75 80
Glu Asp Val Ala Val Tyr Tyr Cys Gln Gln His Tyr Asn Thr Pro Leu
85 90 95
Thr Phe Gly Gly Gly Thr Lys Leu Glu Ile Lys
100 105
<210> 28
<211> 118
<212> PRT
<213> Artificial (Artifical)
<220>
<223> heavy chain variable region of humanized antibody binding to PD-L1
<400> 28
Gln Val Gln Leu Val Gln Ser Gly Ser Glu Leu Lys Lys Pro Gly Ala
1 5 10 15
Ser Val Lys Val Ser Cys Lys Ala Ser Gly Tyr Thr Phe Thr Ser Tyr
20 25 30
Ser Ile Asn Trp Val Arg Gln Ala Pro Gly Lys Gly Leu Glu Trp Val
35 40 45
Ala Tyr Phe Tyr Val Gly Asn Gly Tyr Thr Asp Tyr Asn Glu Lys Phe
50 55 60
Lys Gly Arg Val Thr Ile Thr Ala Asp Glu Ser Thr Ser Thr Ala Tyr
65 70 75 80
Met Glu Leu Ser Ser Leu Arg Ser Glu Asp Thr Ala Val Tyr Tyr Cys
85 90 95
Ala Arg Gly Gly Leu Pro Tyr Tyr Phe Asp Tyr Trp Gly Gln Gly Thr
100 105 110
Leu Val Thr Val Ser Ser
115
<210> 29
<211> 107
<212> PRT
<213> Artificial (Artifical)
<220>
<223> light chain variable region of humanized antibody binding to PD-L1
<400> 29
Asp Ile Gln Met Thr Gln Ser Pro Ser Thr Leu Ser Ala Ser Val Gly
1 5 10 15
Asp Arg Val Thr Ile Thr Cys Lys Thr Ser Gln Asp Val Asn Thr Ala
20 25 30
Val Ala Trp Tyr Gln Gln Lys Pro Gly Lys Ala Pro Lys Leu Leu Leu
35 40 45
Tyr Trp Ala Ser Thr Arg His Thr Gly Ile Pro Ala Arg Phe Ser Gly
50 55 60
Ser Gly Ser Gly Thr Glu Phe Thr Leu Thr Ile Ser Ser Leu Gln Ser
65 70 75 80
Glu Asp Phe Ala Val Tyr Tyr Cys Gln Gln His Tyr Asn Thr Pro Leu
85 90 95
Thr Phe Gly Gly Gly Thr Lys Leu Glu Ile Lys
100 105
<210> 30
<211> 118
<212> PRT
<213> Artificial (Artifical)
<220>
<223> heavy chain variable region of humanized antibody binding to PD-L1
<400> 30
Glu Val Gln Leu Val Gln Ser Gly Ala Glu Val Lys Lys Pro Gly Glu
1 5 10 15
Ser Leu Lys Ile Ser Cys Lys Gly Ser Gly Tyr Ser Phe Thr Ser Tyr
20 25 30
Ser Ile Asn Trp Val Arg Gln Ala Pro Gly Gln Ala Leu Glu Trp Met
35 40 45
Gly Tyr Phe Tyr Val Gly Asn Gly Tyr Thr Asp Tyr Asn Glu Lys Phe
50 55 60
Lys Gly Arg Val Thr Ile Thr Arg Asp Thr Ser Ala Ser Thr Ala Tyr
65 70 75 80
Met Glu Leu Ser Ser Leu Arg Ser Glu Asp Thr Ala Val Tyr Tyr Cys
85 90 95
Ala Arg Gly Asp Leu Pro Phe Tyr Phe Asp Tyr Trp Gly Asp Gly Ile
100 105 110
Leu Val Thr Val Ala Ser
115
<210> 31
<211> 107
<212> PRT
<213> Artificial (Artifical)
<220>
<223> light chain variable region of humanized antibody binding to PD-L1
<400> 31
Asp Ile Val Met Thr Gln Ser Pro Asp Ser Leu Ala Val Ser Leu Gly
1 5 10 15
Glu Arg Ala Thr Ile Asn Cys Lys Thr Ser Gln Asp Val Asn Thr Ala
20 25 30
Val Ala Trp Tyr Gln Gln Lys Pro Gly Gln Pro Pro Lys Leu Leu Ile
35 40 45
Tyr Trp Ala Ser Thr Arg His Thr Gly Val Pro Asp Arg Phe Ser Gly
50 55 60
Ser Gly Ser Gly Thr Asp Phe Thr Leu Lys Ile Ser Arg Val Glu Ala
65 70 75 80
Glu Asp Val Gly Val Tyr Tyr Cys Gln Gln His Tyr Asn Thr Pro Leu
85 90 95
Thr Phe Gly Gly Gly Thr Lys Leu Glu Ile Lys
100 105
<210> 32
<211> 118
<212> PRT
<213> Artificial (Artifical)
<220>
<223> heavy chain variable region of humanized antibody binding to PD-L1
<400> 32
Glu Val Gln Leu Val Gln Ser Gly Ala Glu Val Lys Lys Pro Gly Glu
1 5 10 15
Ser Leu Lys Ile Ser Cys Lys Gly Ser Gly Tyr Ser Phe Thr Ser Tyr
20 25 30
Ser Ile Asn Trp Val Arg Gln Ala Pro Gly Lys Gly Leu Glu Trp Met
35 40 45
Gly Tyr Phe Tyr Val Gly Asn Gly Tyr Thr Asp Tyr Asn Glu Lys Phe
50 55 60
Lys Gly Arg Val Thr Met Thr Arg Asn Thr Ser Ile Ser Thr Ala Tyr
65 70 75 80
Met Glu Leu Ser Ser Leu Arg Ser Glu Asp Thr Ala Val Tyr Tyr Cys
85 90 95
Ala Arg Gly Gly Leu Pro Tyr Tyr Phe Asp Tyr Trp Gly Gln Gly Thr
100 105 110
Leu Val Thr Val Ser Ser
115
<210> 33
<211> 107
<212> PRT
<213> Artificial (Artifical)
<220>
<223> light chain variable region of humanized antibody binding to PD-L1
<400> 33
Asp Ile Gln Met Thr Gln Ser Pro Ser Ser Val Ser Ala Ser Val Gly
1 5 10 15
Asp Arg Val Thr Ile Thr Cys Lys Thr Ser Gln Asp Val Asn Thr Ala
20 25 30
Val Ala Trp Tyr Gln Gln Lys Pro Gly Lys Ala Pro Lys Leu Leu Leu
35 40 45
Tyr Trp Ala Ser Thr Arg His Thr Gly Val Pro Ser Arg Phe Ser Gly
50 55 60
Ser Gly Ser Gly Thr Asp Phe Thr Leu Thr Ile Ser Ser Leu Gln Pro
65 70 75 80
Glu Asp Val Ala Thr Tyr Tyr Cys Gln Gln His Tyr Asn Thr Pro Leu
85 90 95
Thr Phe Gly Gly Gly Thr Lys Leu Glu Ile Lys
100 105
<210> 34
<211> 118
<212> PRT
<213> Artificial (Artifical)
<220>
<223> heavy chain variable region of humanized antibody binding to PD-L1
<400> 34
Glu Val Gln Leu Val Gln Ser Gly Ala Glu Val Lys Lys Pro Gly Glu
1 5 10 15
Ser Leu Lys Ile Ser Cys Lys Gly Ser Gly Tyr Ser Phe Thr Ser Tyr
20 25 30
Ser Ile Asn Trp Val Arg Gln Ala Pro Gly Lys Gly Leu Glu Trp Met
35 40 45
Gly Tyr Phe Tyr Val Gly Asn Gly Tyr Thr Asp Tyr Asn Glu Lys Phe
50 55 60
Lys Gly Arg Val Thr Met Thr Arg Asn Thr Ser Ile Ser Thr Ala Tyr
65 70 75 80
Met Glu Leu Ser Ser Leu Arg Ser Glu Asp Thr Ala Val Tyr Tyr Cys
85 90 95
Ala Arg Gly Gly Leu Pro Tyr Tyr Phe Asp Tyr Trp Gly Gln Gly Thr
100 105 110
Leu Val Thr Val Ser Ser
115
<210> 35
<211> 107
<212> PRT
<213> Artificial (Artifical)
<220>
<223> light chain variable region of humanized antibody binding to PD-L1
<400> 35
Asp Ile Gln Met Thr Gln Ser Pro Ser Ser Val Ser Ala Ser Val Gly
1 5 10 15
Asp Arg Val Thr Ile Thr Cys Lys Thr Ser Gln Asp Val Asn Thr Ala
20 25 30
Val Ala Trp Tyr Gln Gln Lys Pro Gly Gln Pro Pro Lys Leu Leu Ile
35 40 45
Tyr Trp Ala Ser Thr Arg His Thr Gly Val Pro Asp Arg Phe Ser Gly
50 55 60
Ser Gly Ser Gly Thr Asp Phe Thr Leu Thr Ile Ser Ser Leu Gln Ala
65 70 75 80
Glu Asp Val Ala Val Tyr Tyr Cys Gln Gln His Tyr Asn Thr Pro Leu
85 90 95
Thr Phe Gly Gly Gly Thr Lys Leu Glu Ile Lys
100 105
<210> 36
<211> 118
<212> PRT
<213> Artificial (Artifical)
<220>
<223> heavy chain variable region of humanized antibody binding to PD-L1
<400> 36
Glu Val Gln Leu Val Gln Ser Gly Ala Glu Val Lys Lys Pro Gly Glu
1 5 10 15
Ser Leu Lys Ile Ser Cys Lys Gly Ser Gly Tyr Ser Phe Thr Ser Tyr
20 25 30
Ser Ile Asn Trp Val Arg Gln Ala Pro Gly Lys Gly Leu Glu Trp Met
35 40 45
Gly Tyr Phe Tyr Val Gly Asn Gly Tyr Thr Asp Tyr Asn Glu Lys Phe
50 55 60
Lys Gly Arg Val Thr Ile Thr Arg Asp Thr Ser Ala Ser Thr Ala Tyr
65 70 75 80
Met Glu Leu Ser Ser Leu Arg Ser Glu Asp Thr Ala Val Tyr Tyr Cys
85 90 95
Ala Arg Gly Gly Leu Pro Tyr Tyr Phe Asp Tyr Trp Gly Gln Gly Thr
100 105 110
Leu Val Thr Val Ser Ser
115
<210> 37
<211> 107
<212> PRT
<213> Artificial (Artifical)
<220>
<223> light chain variable region of humanized antibody binding to PD-L1
<400> 37
Asp Ile Gln Leu Thr Gln Ser Pro Ser Phe Leu Ser Ala Ser Val Gly
1 5 10 15
Asp Arg Val Thr Ile Thr Cys Lys Thr Ser Gln Asp Val Asn Thr Ala
20 25 30
Val Ala Trp Tyr Gln Gln Lys Pro Gly Gln Pro Pro Lys Leu Leu Ile
35 40 45
Tyr Trp Ala Ser Thr Arg His Thr Gly Val Pro Ser Arg Phe Ser Gly
50 55 60
Ser Gly Ser Gly Thr Asp Phe Thr Leu Thr Ile Ser Ser Leu Gln Pro
65 70 75 80
Glu Asp Phe Ala Thr Tyr Tyr Cys Gln Gln His Tyr Asn Thr Pro Leu
85 90 95
Thr Phe Gly Gly Gly Thr Lys Leu Glu Ile Lys
100 105
<210> 38
<211> 118
<212> PRT
<213> Artificial (Artifical)
<220>
<223> heavy chain variable region of humanized antibody binding to PD-L1
<400> 38
Gln Val Gln Leu Val Gln Ser Gly Ser Glu Leu Lys Lys Pro Gly Ala
1 5 10 15
Ser Val Lys Val Ser Cys Lys Ala Ser Gly Tyr Thr Phe Thr Ser Tyr
20 25 30
Ser Ile Asn Trp Val Arg Gln Ala Pro Gly Lys Gly Leu Glu Trp Val
35 40 45
Ala Tyr Phe Tyr Val Gly Asn Gly Tyr Thr Asp Tyr Asn Glu Lys Phe
50 55 60
Lys Gly Arg Val Thr Ile Thr Ala Asp Glu Ser Thr Ser Thr Ala Tyr
65 70 75 80
Met Glu Leu Ser Ser Leu Arg Ser Glu Asp Thr Ala Val Tyr Tyr Cys
85 90 95
Ala Arg Gly Gly Leu Pro Tyr Tyr Phe Asp Tyr Trp Gly Gln Gly Thr
100 105 110
Leu Val Thr Val Ser Ser
115
<210> 39
<211> 107
<212> PRT
<213> Artificial (Artifical)
<220>
<223> light chain variable region of humanized antibody binding to PD-L1
<400> 39
Glu Ile Val Met Thr Gln Ser Pro Ala Thr Leu Ser Val Ser Pro Gly
1 5 10 15
Glu Arg Ala Thr Leu Ser Cys Lys Thr Ser Gln Asp Val Asn Thr Ala
20 25 30
Val Ala Trp Tyr Leu Gln Lys Pro Gly Gln Ser Pro Gln Leu Leu Ile
35 40 45
Tyr Trp Ala Ser Thr Arg His Thr Gly Val Pro Ser Arg Phe Ser Gly
50 55 60
Ser Gly Ser Gly Thr Asp Tyr Thr Leu Thr Ile Ser Ser Leu Gln Pro
65 70 75 80
Glu Asp Phe Ala Thr Tyr Tyr Cys Gln Gln His Tyr Asn Thr Pro Leu
85 90 95
Thr Phe Gly Gly Gly Thr Lys Leu Glu Ile Lys
100 105
<210> 40
<211> 118
<212> PRT
<213> Artificial (Artifical)
<220>
<223> heavy chain variable region of humanized antibody binding to PD-L1
<400> 40
Gln Val Gln Leu Val Gln Ser Gly Ser Glu Leu Lys Lys Pro Gly Ala
1 5 10 15
Ser Val Lys Val Ser Cys Lys Ala Ser Gly Tyr Thr Phe Thr Ser Tyr
20 25 30
Ser Ile Asn Trp Val Arg Gln Ala Pro Gly Lys Gly Leu Glu Trp Met
35 40 45
Gly Tyr Phe Tyr Val Gly Asn Gly Tyr Thr Asp Tyr Asn Glu Lys Phe
50 55 60
Lys Gly Arg Val Thr Ile Thr Arg Asp Thr Ser Ala Ser Thr Ala Tyr
65 70 75 80
Met Glu Leu Ser Ser Leu Arg Ser Glu Asp Thr Ala Val Tyr Tyr Cys
85 90 95
Ala Arg Gly Gly Leu Pro Tyr Tyr Phe Asp Tyr Trp Gly Gln Gly Thr
100 105 110
Leu Val Thr Val Ser Ser
115
<210> 41
<211> 107
<212> PRT
<213> Artificial (Artifical)
<220>
<223> light chain variable region of humanized antibody binding to PD-L1
<400> 41
Asp Ile Gln Met Thr Gln Ser Pro Ser Ser Val Ser Ala Ser Val Gly
1 5 10 15
Asp Arg Val Thr Ile Thr Cys Lys Thr Ser Gln Asp Val Asn Thr Ala
20 25 30
Val Ala Trp Tyr Gln Gln Lys Pro Asp Gln Ser Pro Lys Leu Leu Ile
35 40 45
Lys Trp Ala Ser Thr Arg His Thr Gly Val Pro Ser Arg Phe Ser Gly
50 55 60
Ser Gly Ser Gly Thr Asp Phe Thr Leu Thr Ile Ser Ser Leu Gln Pro
65 70 75 80
Glu Asp Phe Ala Thr Tyr Tyr Cys Gln Gln His Tyr Asn Thr Pro Leu
85 90 95
Thr Phe Gly Gly Gly Thr Lys Leu Glu Ile Lys
100 105
<210> 42
<211> 118
<212> PRT
<213> Artificial (Artifical)
<220>
<223> heavy chain variable region of humanized antibody binding to PD-L1
<400> 42
Gln Met Gln Leu Val Gln Ser Gly Ala Glu Val Lys Lys Thr Gly Ser
1 5 10 15
Ser Val Lys Val Ser Cys Lys Ala Ser Gly Tyr Thr Phe Thr Ser Tyr
20 25 30
Ser Ile Asn Trp Val Arg Gln Met Pro Gly Lys Gly Leu Glu Trp Met
35 40 45
Gly Tyr Phe Tyr Val Gly Asn Gly Tyr Thr Asp Tyr Asn Glu Lys Phe
50 55 60
Lys Gly Arg Val Thr Ile Thr Ala Asp Glu Ser Thr Ser Thr Ala Tyr
65 70 75 80
Met Glu Leu Ser Ser Leu Arg Ser Glu Asp Thr Ala Val Tyr Tyr Cys
85 90 95
Ala Arg Gly Gly Leu Pro Tyr Tyr Phe Asp Tyr Trp Gly Gln Gly Thr
100 105 110
Leu Val Thr Val Ser Ser
115
<210> 43
<211> 107
<212> PRT
<213> Artificial (Artifical)
<220>
<223> light chain variable region of humanized antibody binding to PD-L1
<400> 43
Asp Ile Gln Leu Thr Gln Ser Pro Ser Phe Leu Ser Ala Ser Val Gly
1 5 10 15
Asp Arg Val Thr Ile Thr Cys Lys Thr Ser Gln Asp Val Asn Thr Ala
20 25 30
Val Ala Trp Tyr Gln Gln Lys Pro Gly Lys Ala Pro Lys Leu Leu Ile
35 40 45
Tyr Trp Ala Ser Thr Arg His Thr Gly Val Pro Ser Arg Phe Ser Gly
50 55 60
Ser Gly Ser Gly Thr Asp Phe Thr Leu Thr Ile Ser Ser Leu Gln Pro
65 70 75 80
Glu Asp Val Ala Thr Tyr Tyr Cys Gln Gln His Tyr Asn Thr Pro Leu
85 90 95
Thr Phe Gly Gly Gly Thr Lys Leu Glu Ile Lys
100 105
<210> 44
<211> 118
<212> PRT
<213> Artificial (Artifical)
<220>
<223> heavy chain variable region of humanized antibody binding to PD-L1
<400> 44
Gln Met Gln Leu Val Gln Ser Gly Ala Glu Val Lys Lys Thr Gly Ser
1 5 10 15
Ser Val Lys Val Ser Cys Lys Ala Ser Gly Tyr Thr Phe Thr Ser Tyr
20 25 30
Ser Ile Asn Trp Val Arg Gln Ala Arg Gly Gln Arg Leu Glu Trp Ile
35 40 45
Gly Tyr Phe Tyr Val Gly Asn Gly Tyr Thr Asp Tyr Asn Glu Lys Phe
50 55 60
Lys Gly Arg Val Thr Ile Thr Ala Asp Glu Ser Thr Ser Thr Ala Tyr
65 70 75 80
Met Glu Leu Ser Ser Leu Arg Ser Glu Asp Thr Ala Val Tyr Tyr Cys
85 90 95
Ala Arg Gly Gly Leu Pro Tyr Tyr Phe Asp Tyr Trp Gly Gln Gly Thr
100 105 110
Leu Val Thr Val Ser Ser
115
<210> 45
<211> 107
<212> PRT
<213> Artificial (Artifical)
<220>
<223> light chain variable region of humanized antibody binding to PD-L1
<400> 45
Asp Ile Gln Met Thr Gln Ser Pro Ser Ser Leu Ser Ala Ser Val Gly
1 5 10 15
Asp Arg Val Thr Ile Thr Cys Lys Thr Ser Gln Asp Val Asn Thr Ala
20 25 30
Val Ala Trp Tyr Gln Gln Lys Pro Asp Gln Ser Pro Lys Leu Leu Ile
35 40 45
Lys Trp Ala Ser Thr Arg His Thr Gly Ile Pro Ala Arg Phe Ser Gly
50 55 60
Ser Gly Ser Gly Thr Glu Phe Thr Leu Thr Ile Ser Ser Leu Gln Ser
65 70 75 80
Glu Asp Phe Ala Val Tyr Tyr Cys Gln Gln His Tyr Asn Thr Pro Leu
85 90 95
Thr Phe Gly Gly Gly Thr Lys Leu Glu Ile Lys
100 105
<210> 46
<211> 118
<212> PRT
<213> Artificial (Artifical)
<220>
<223> heavy chain variable region of humanized antibody binding to PD-L1
<400> 46
Glu Val Gln Leu Val Gln Ser Gly Ala Glu Val Lys Lys Pro Gly Glu
1 5 10 15
Ser Leu Lys Ile Ser Cys Lys Gly Ser Gly Tyr Ser Phe Thr Ser Tyr
20 25 30
Ser Ile Asn Trp Val Arg Gln Ala Pro Gly Lys Gly Leu Glu Trp Met
35 40 45
Gly Tyr Phe Tyr Val Gly Asn Gly Tyr Thr Asp Tyr Asn Glu Lys Phe
50 55 60
Lys Gly Arg Val Thr Ile Thr Thr Asp Glu Ser Thr Ser Thr Ala Tyr
65 70 75 80
Met Glu Leu Ser Ser Leu Arg Ser Glu Asp Thr Ala Val Tyr Tyr Cys
85 90 95
Ala Arg Gly Gly Leu Pro Tyr Tyr Phe Asp Tyr Trp Gly Gln Gly Thr
100 105 110
Leu Val Thr Val Ser Ser
115
<210> 47
<211> 107
<212> PRT
<213> Artificial (Artifical)
<220>
<223> light chain variable region of humanized antibody binding to PD-L1
<400> 47
Asp Ile Gln Leu Thr Gln Ser Pro Ser Ser Leu Ser Ala Ser Val Gly
1 5 10 15
Asp Arg Val Thr Ile Thr Cys Lys Thr Ser Gln Asp Val Asn Thr Ala
20 25 30
Val Ala Trp Tyr Gln Gln Lys Pro Asp Gln Ser Pro Lys Leu Leu Ile
35 40 45
Lys Trp Ala Ser Thr Arg His Thr Gly Val Pro Ser Arg Phe Ser Gly
50 55 60
Ser Gly Ser Gly Thr Asp Phe Thr Leu Thr Ile Ser Ser Leu Gln Pro
65 70 75 80
Glu Asp Phe Ala Thr Tyr Tyr Cys Gln Gln His Tyr Asn Thr Pro Leu
85 90 95
Thr Phe Gly Gly Gly Thr Lys Leu Glu Ile Lys
100 105
<210> 48
<211> 106
<212> PRT
<213> Homo sapiens (Homo sapiens)
<400> 48
Thr Val Ala Ala Pro Ser Val Phe Ile Phe Pro Pro Ser Asp Glu Gln
1 5 10 15
Leu Lys Ser Gly Thr Ala Ser Val Val Cys Leu Leu Asn Asn Phe Tyr
20 25 30
Pro Arg Glu Ala Lys Val Gln Trp Lys Val Asp Asn Ala Leu Gln Ser
35 40 45
Gly Asn Ser Gln Glu Ser Val Thr Glu Gln Asp Ser Lys Asp Ser Thr
50 55 60
Tyr Ser Leu Ser Ser Thr Leu Thr Leu Ser Lys Ala Asp Tyr Glu Lys
65 70 75 80
His Lys Val Tyr Ala Cys Glu Val Thr His Gln Gly Leu Ser Ser Pro
85 90 95
Val Thr Lys Ser Phe Asn Arg Gly Glu Cys
100 105
<210> 49
<211> 105
<212> PRT
<213> Homo sapiens (Homo sapiens)
<400> 49
Gln Pro Lys Ala Ala Pro Ser Val Thr Leu Phe Pro Pro Ser Ser Glu
1 5 10 15
Glu Leu Gln Ala Asn Lys Ala Thr Leu Val Cys Leu Ile Ser Asp Phe
20 25 30
Tyr Pro Gly Ala Val Thr Val Ala Trp Lys Ala Asp Ser Ser Pro Val
35 40 45
Lys Ala Gly Val Glu Thr Thr Thr Pro Ser Lys Gln Ser Asn Asn Lys
50 55 60
Tyr Ala Ala Ser Ser Tyr Leu Ser Leu Thr Pro Glu Gln Trp Lys Ser
65 70 75 80
His Arg Ser Tyr Ser Cys Gln Val Thr His Glu Gly Ser Thr Val Glu
85 90 95
Lys Thr Val Ala Pro Thr Glu Cys Ser
100 105
<210> 50
<211> 328
<212> PRT
<213> Homo sapiens (Homo sapiens)
<400> 50
Thr Lys Gly Pro Ser Val Phe Pro Leu Ala Pro Ser Ser Lys Ser Thr
1 5 10 15
Ser Gly Gly Thr Ala Ala Leu Gly Cys Leu Val Lys Asp Tyr Phe Pro
20 25 30
Glu Pro Val Thr Val Ser Trp Asn Ser Gly Ala Leu Thr Ser Gly Val
35 40 45
His Thr Phe Pro Ala Val Leu Gln Ser Ser Gly Leu Tyr Ser Leu Ser
50 55 60
Ser Val Val Thr Val Pro Ser Ser Ser Leu Gly Thr Gln Thr Tyr Ile
65 70 75 80
Cys Asn Val Asn His Lys Pro Ser Asn Thr Lys Val Asp Lys Lys Val
85 90 95
Glu Pro Lys Ser Cys Asp Lys Thr His Thr Cys Pro Pro Cys Pro Ala
100 105 110
Pro Glu Leu Leu Gly Gly Pro Ser Val Phe Leu Phe Pro Pro Lys Pro
115 120 125
Lys Asp Thr Leu Met Ile Ser Arg Thr Pro Glu Val Thr Cys Val Val
130 135 140
Val Asp Val Ser His Glu Asp Pro Glu Val Lys Phe Asn Trp Tyr Val
145 150 155 160
Asp Gly Val Glu Val His Asn Ala Lys Thr Lys Pro Arg Glu Glu Gln
165 170 175
Tyr Asn Ser Thr Tyr Arg Val Val Ser Val Leu Thr Val Leu His Gln
180 185 190
Asp Trp Leu Asn Gly Lys Glu Tyr Lys Cys Lys Val Ser Asn Lys Ala
195 200 205
Leu Pro Ala Pro Ile Glu Lys Thr Ile Ser Lys Ala Lys Gly Gln Pro
210 215 220
Arg Glu Pro Gln Val Tyr Thr Leu Pro Pro Ser Arg Asp Glu Leu Thr
225 230 235 240
Lys Asn Gln Val Ser Leu Thr Cys Leu Val Lys Gly Phe Tyr Pro Ser
245 250 255
Asp Ile Ala Val Glu Trp Glu Ser Asn Gly Gln Pro Glu Asn Asn Tyr
260 265 270
Lys Thr Thr Pro Pro Val Leu Asp Ser Asp Gly Ser Phe Phe Leu Tyr
275 280 285
Ser Lys Leu Thr Val Asp Lys Ser Arg Trp Gln Gln Gly Asn Val Phe
290 295 300
Ser Cys Ser Val Met His Glu Ala Leu His Asn His Tyr Thr Gln Lys
305 310 315 320
Ser Leu Ser Leu Ser Pro Gly Lys
325
<210> 51
<211> 464
<212> PRT
<213> Artificial (Artifical)
<220>
<223> heavy chain of chimeric antibody binding to PD-L1
<400> 51
Met Gly Trp Ser Trp Ile Leu Leu Phe Leu Leu Ser Val Thr Ala Gly
1 5 10 15
Val His Ser Glu Val Gln Leu Gln Gln Ser Gly Ala Glu Leu Val Arg
20 25 30
Pro Gly Ser Ser Val Lys Met Ser Cys Lys Thr Ser Gly Tyr Thr Phe
35 40 45
Thr Ser Tyr Ser Ile Asn Trp Val Lys Gln Arg Pro Gly Gln Gly Leu
50 55 60
Glu Trp Ile Ala Tyr Phe Tyr Val Gly Asn Gly Tyr Thr Asp Tyr Asn
65 70 75 80
Glu Lys Phe Lys Gly Lys Ala Thr Leu Thr Ser Asp Thr Ser Ser Ser
85 90 95
Thr Ala Tyr Met Gln Leu Ser Ser Leu Thr Ser Glu Asp Ser Ala Ile
100 105 110
Tyr Phe Cys Ala Arg Gly Gly Leu Pro Tyr Tyr Phe Asp Tyr Trp Gly
115 120 125
Gln Gly Thr Thr Leu Thr Val Ser Ser Ala Ser Thr Lys Gly Pro Ser
130 135 140
Val Phe Pro Leu Ala Pro Cys Ser Arg Ser Thr Ser Glu Ser Thr Ala
145 150 155 160
Ala Leu Gly Cys Leu Val Lys Asp Tyr Phe Pro Glu Pro Val Thr Val
165 170 175
Ser Trp Asn Ser Gly Ala Leu Thr Ser Gly Val His Thr Phe Pro Ala
180 185 190
Val Leu Gln Ser Ser Gly Leu Tyr Ser Leu Ser Ser Val Val Thr Val
195 200 205
Pro Ser Ser Ser Leu Gly Thr Lys Thr Tyr Thr Cys Asn Val Asp His
210 215 220
Lys Pro Ser Asn Thr Lys Val Asp Lys Arg Val Glu Ser Lys Tyr Gly
225 230 235 240
Pro Pro Cys Pro Pro Cys Pro Ala Pro Glu Phe Leu Gly Gly Pro Ser
245 250 255
Val Phe Leu Phe Pro Pro Lys Pro Lys Asp Thr Leu Met Ile Ser Arg
260 265 270
Thr Pro Glu Val Thr Cys Val Val Val Asp Val Ser Gln Glu Asp Pro
275 280 285
Glu Val Gln Phe Asn Trp Tyr Val Asp Gly Val Glu Val His Asn Ala
290 295 300
Lys Thr Lys Pro Arg Glu Glu Gln Phe Asn Ser Thr Tyr Arg Val Val
305 310 315 320
Ser Val Leu Thr Val Leu His Gln Asp Trp Leu Asn Gly Lys Glu Tyr
325 330 335
Lys Cys Lys Val Ser Asn Lys Gly Leu Pro Ser Ser Ile Glu Lys Thr
340 345 350
Ile Ser Lys Ala Lys Gly Gln Pro Arg Glu Pro Gln Val Tyr Thr Leu
355 360 365
Pro Pro Ser Gln Glu Glu Met Thr Lys Asn Gln Val Ser Leu Thr Cys
370 375 380
Leu Val Lys Gly Phe Tyr Pro Ser Asp Ile Ala Val Glu Trp Glu Ser
385 390 395 400
Asn Gly Gln Pro Glu Asn Asn Tyr Lys Thr Thr Pro Pro Val Leu Asp
405 410 415
Ser Asp Gly Ser Phe Phe Leu Tyr Ser Arg Leu Thr Val Asp Lys Ser
420 425 430
Arg Trp Gln Glu Gly Asn Val Phe Ser Cys Ser Val Met His Glu Ala
435 440 445
Leu His Asn His Tyr Thr Gln Lys Ser Leu Ser Leu Ser Leu Gly Lys
450 455 460
<210> 52
<211> 1392
<212> DNA
<213> Artificial (Artifical)
<220>
<223> heavy chain of chimeric antibody binding to PD-L1
<400> 52
atgggctgga gctggatcct gctgttcctc ctgagcgtga cagcaggagt gcacagcgag 60
gtgcagctgc agcagagcgg ggcagaactg gtgcggcctg ggtcaagcgt gaagatgtca 120
tgtaaaacaa gcggctatac tttcacatcc tactctatca actgggtgaa gcagaggcca 180
ggacagggac tggagtggat cgcctacttc tatgtgggca acggctacac cgactataat 240
gagaagttta agggcaaggc caccctgaca agcgatacaa gctcctctac cgcctatatg 300
cagctgagct ccctgaccag cgaggactcc gccatctact tctgcgcaag gggaggactg 360
ccatactatt ttgattattg gggccagggc accacactga cagtgtctag cgccagcacc 420
aagggaccat ccgtgttccc actggcacca tgcagcagat ccacatctga gagcaccgcc 480
gccctgggat gtctggtgaa ggactacttc cctgagccag tgaccgtgtc ttggaatagc 540
ggcgccctga caagcggagt gcacaccttt cctgccgtgc tgcagtcctc tggcctgtac 600
tccctgagct ccgtggtgac agtgccctct agctccctgg gcaccaagac atatacctgc 660
aacgtggacc acaagccttc taataccaag gtggataaga gggtggagag caagtacgga 720
ccaccttgcc caccatgtcc agcacctgag tttctgggag gaccaagcgt gttcctgttt 780
cctccaaagc ctaaggacac actgatgatc agccgcacac ctgaggtgac ctgcgtggtg 840
gtggacgtgt cccaggagga tccagaggtg cagttcaact ggtacgtgga tggcgtggag 900
gtgcacaatg ccaagaccaa gcctagggag gagcagttta actctacata ccgcgtggtg 960
agcgtgctga ccgtgctgca ccaggattgg ctgaacggca aggagtataa gtgcaaggtg 1020
tctaataagg gcctgccatc tagcatcgag aagacaatct ccaaggcaaa gggacagcct 1080
agggagccac aggtgtacac cctgccccct tcccaggagg agatgacaaa gaaccaggtg 1140
tctctgacct gtctggtgaa gggcttctat ccaagcgaca tcgccgtgga gtgggagtcc 1200
aatggccagc ccgagaacaa ttacaagacc acaccacccg tgctggactc cgatggctct 1260
ttctttctgt attcccggct gaccgtggat aagtctagat ggcaggaggg caacgtgttc 1320
agctgttctg tgatgcacga agcactgcac aaccattaca ctcagaagtc cctgtccctg 1380
tccctgggca aa 1392
<210> 53
<211> 233
<212> PRT
<213> Artificial (Artifical)
<220>
<223> light chain of chimeric antibody binding to PD-L1
<400> 53
Met Gly Trp Ser Trp Ile Leu Leu Phe Leu Leu Ser Val Thr Ala Gly
1 5 10 15
Val His Ser Asp Ile Val Met Thr Gln Ser His Lys Phe Met Ser Thr
20 25 30
Ser Val Gly Asp Arg Val Ser Ile Thr Cys Lys Thr Ser Gln Asp Val
35 40 45
Asn Thr Ala Val Ala Trp Tyr Gln Gln Lys Pro Gly His Ser Pro Lys
50 55 60
Leu Leu Phe Tyr Trp Ala Ser Thr Arg His Thr Gly Val Pro Asp Arg
65 70 75 80
Phe Thr Gly Gly Gly Ser Gly Thr Asp Tyr Thr Leu Thr Ile Ser Ser
85 90 95
Val Gln Ala Glu Asp Leu Ala Leu Tyr Tyr Cys Gln Gln His Tyr Asn
100 105 110
Thr Pro Leu Thr Phe Gly Ala Gly Thr Arg Leu Glu Leu Lys Arg Thr
115 120 125
Val Ala Ala Pro Ser Val Phe Ile Phe Pro Pro Ser Asp Glu Gln Leu
130 135 140
Lys Ser Gly Thr Ala Ser Val Val Cys Leu Leu Asn Asn Phe Tyr Pro
145 150 155 160
Arg Glu Ala Lys Val Gln Trp Lys Val Asp Asn Ala Leu Gln Ser Gly
165 170 175
Asn Ser Gln Glu Ser Val Thr Glu Gln Asp Ser Lys Asp Ser Thr Tyr
180 185 190
Ser Leu Ser Ser Thr Leu Thr Leu Ser Lys Ala Asp Tyr Glu Lys His
195 200 205
Lys Val Tyr Ala Cys Glu Val Thr His Gln Gly Leu Ser Ser Pro Val
210 215 220
Thr Lys Ser Phe Asn Arg Gly Glu Cys
225 230
<210> 54
<211> 699
<212> DNA
<213> Artificial (Artifical)
<220>
<223> light chain of chimeric antibody binding to PD-L1
<400> 54
atgggctgga gctggatcct gctgttcctc ctgagcgtga cagcaggagt gcacagcgat 60
atcgtgatga cccagagcca caagtttatg agcaccagcg tcggagacag agtcagtatt 120
acctgcaaaa ccagccagga tgtgaacacc gccgtggcct ggtaccagca gaagccaggc 180
cactctccca agctgctgtt ctattgggca agcaccaggc acacaggagt gccagaccgc 240
tttaccggag gaggatccgg aacagattac accctgacaa tcagctccgt gcaggcagag 300
gacctggccc tgtactattg ccagcagcac tataataccc ctctgacatt cggcgcagga 360
accaggctgg agctgaagag aacagtggcc gccccaagcg tgttcatctt tcccccttcc 420
gacgagcagc tgaagtccgg caccgcctct gtggtgtgcc tgctgaacaa cttctaccct 480
cgggaggcca aggtgcagtg gaaggtggat aacgccctgc agtccggcaa ttctcaggag 540
agcgtgaccg agcaggactc caaggattct acatatagcc tgtctagcac cctgacactg 600
tctaaggccg attacgagaa gcacaaggtg tatgcctgcg aggtcactca ccaggggctg 660
tcttcaccag tcaccaagtc cttcaatcgg ggggaatgc 699
<210> 55
<211> 464
<212> PRT
<213> Artificial (Artifical)
<220>
<223> heavy chain-19 aa leader sequence of humanized antibody binding to PD-L1
<400> 55
Met Gly Trp Ser Trp Ile Leu Leu Phe Leu Leu Ser Val Thr Ala Gly
1 5 10 15
Val His Ser Glu Val Gln Leu Val Gln Ser Gly Ala Glu Val Lys Lys
20 25 30
Pro Gly Glu Ser Leu Lys Ile Ser Cys Lys Gly Ser Gly Tyr Ser Phe
35 40 45
Thr Ser Tyr Ser Ile Asn Trp Val Arg Gln Met Pro Gly Lys Gly Leu
50 55 60
Glu Trp Met Gly Tyr Phe Tyr Val Gly Asn Gly Tyr Thr Asp Tyr Asn
65 70 75 80
Glu Lys Phe Lys Gly Arg Val Thr Met Thr Arg Asn Thr Ser Ile Ser
85 90 95
Thr Ala Tyr Met Glu Leu Ser Ser Leu Arg Ser Glu Asp Thr Ala Val
100 105 110
Tyr Tyr Cys Ala Arg Gly Gly Leu Pro Tyr Tyr Phe Asp Tyr Trp Gly
115 120 125
Gln Gly Thr Leu Val Thr Val Ser Ser Ala Ser Thr Lys Gly Pro Ser
130 135 140
Val Phe Pro Leu Ala Pro Cys Ser Arg Ser Thr Ser Glu Ser Thr Ala
145 150 155 160
Ala Leu Gly Cys Leu Val Lys Asp Tyr Phe Pro Glu Pro Val Thr Val
165 170 175
Ser Trp Asn Ser Gly Ala Leu Thr Ser Gly Val His Thr Phe Pro Ala
180 185 190
Val Leu Gln Ser Ser Gly Leu Tyr Ser Leu Ser Ser Val Val Thr Val
195 200 205
Pro Ser Ser Ser Leu Gly Thr Lys Thr Tyr Thr Cys Asn Val Asp His
210 215 220
Lys Pro Ser Asn Thr Lys Val Asp Lys Arg Val Glu Ser Lys Tyr Gly
225 230 235 240
Pro Pro Cys Pro Pro Cys Pro Ala Pro Glu Phe Leu Gly Gly Pro Ser
245 250 255
Val Phe Leu Phe Pro Pro Lys Pro Lys Asp Thr Leu Met Ile Ser Arg
260 265 270
Thr Pro Glu Val Thr Cys Val Val Val Asp Val Ser Gln Glu Asp Pro
275 280 285
Glu Val Gln Phe Asn Trp Tyr Val Asp Gly Val Glu Val His Asn Ala
290 295 300
Lys Thr Lys Pro Arg Glu Glu Gln Phe Asn Ser Thr Tyr Arg Val Val
305 310 315 320
Ser Val Leu Thr Val Leu His Gln Asp Trp Leu Asn Gly Lys Glu Tyr
325 330 335
Lys Cys Lys Val Ser Asn Lys Gly Leu Pro Ser Ser Ile Glu Lys Thr
340 345 350
Ile Ser Lys Ala Lys Gly Gln Pro Arg Glu Pro Gln Val Tyr Thr Leu
355 360 365
Pro Pro Ser Gln Glu Glu Met Thr Lys Asn Gln Val Ser Leu Thr Cys
370 375 380
Leu Val Lys Gly Phe Tyr Pro Ser Asp Ile Ala Val Glu Trp Glu Ser
385 390 395 400
Asn Gly Gln Pro Glu Asn Asn Tyr Lys Thr Thr Pro Pro Val Leu Asp
405 410 415
Ser Asp Gly Ser Phe Phe Leu Tyr Ser Arg Leu Thr Val Asp Lys Ser
420 425 430
Arg Trp Gln Glu Gly Asn Val Phe Ser Cys Ser Val Met His Glu Ala
435 440 445
Leu His Asn His Tyr Thr Gln Lys Ser Leu Ser Leu Ser Leu Gly Lys
450 455 460
<210> 56
<211> 233
<212> PRT
<213> Artificial (Artifical)
<220>
<223> light chain-19 aa leader sequence of humanized antibody binding to PD-L1
<400> 56
Met Gly Trp Ser Trp Ile Leu Leu Phe Leu Leu Ser Val Thr Ala Gly
1 5 10 15
Val His Ser Asp Ile Gln Met Thr Gln Ser Pro Ser Thr Leu Ser Ala
20 25 30
Ser Val Gly Asp Arg Val Thr Ile Thr Cys Lys Thr Ser Gln Asp Val
35 40 45
Asn Thr Ala Val Ala Trp Tyr Gln Gln Lys Pro Gly Lys Ala Pro Lys
50 55 60
Leu Leu Ile Tyr Trp Ala Ser Thr Arg His Thr Gly Val Pro Ser Arg
65 70 75 80
Phe Ser Gly Ser Gly Ser Gly Thr Glu Phe Thr Leu Thr Ile Ser Ser
85 90 95
Leu Gln Pro Glu Asp Phe Ala Thr Tyr Tyr Cys Gln Gln His Tyr Asn
100 105 110
Thr Pro Leu Thr Phe Gly Gly Gly Thr Lys Leu Glu Ile Lys Arg Thr
115 120 125
Val Ala Ala Pro Ser Val Phe Ile Phe Pro Pro Ser Asp Glu Gln Leu
130 135 140
Lys Ser Gly Thr Ala Ser Val Val Cys Leu Leu Asn Asn Phe Tyr Pro
145 150 155 160
Arg Glu Ala Lys Val Gln Trp Lys Val Asp Asn Ala Leu Gln Ser Gly
165 170 175
Asn Ser Gln Glu Ser Val Thr Glu Gln Asp Ser Lys Asp Ser Thr Tyr
180 185 190
Ser Leu Ser Ser Thr Leu Thr Leu Ser Lys Ala Asp Tyr Glu Lys His
195 200 205
Lys Val Tyr Ala Cys Glu Val Thr His Gln Gly Leu Ser Ser Pro Val
210 215 220
Thr Lys Ser Phe Asn Arg Gly Glu Cys
225 230
<210> 57
<211> 464
<212> PRT
<213> Artificial (Artifical)
<220>
<223> heavy chain-19 aa leader sequence of humanized antibody binding to PD-L1
<400> 57
Met Gly Trp Ser Trp Ile Leu Leu Phe Leu Leu Ser Val Thr Ala Gly
1 5 10 15
Val His Ser Glu Val Gln Leu Val Gln Ser Gly Ala Glu Val Lys Lys
20 25 30
Pro Gly Glu Ser Leu Lys Ile Ser Cys Lys Gly Ser Gly Tyr Ser Phe
35 40 45
Thr Ser Tyr Ser Ile Asn Trp Val Arg Gln Ala Pro Gly Lys Gly Leu
50 55 60
Glu Trp Met Gly Tyr Phe Tyr Val Gly Asn Gly Tyr Thr Asp Tyr Asn
65 70 75 80
Glu Lys Phe Lys Gly Arg Val Thr Met Thr Arg Asn Thr Ser Ile Ser
85 90 95
Thr Ala Tyr Met Glu Leu Ser Ser Leu Arg Ser Glu Asp Thr Ala Val
100 105 110
Tyr Tyr Cys Ala Arg Gly Gly Leu Pro Tyr Tyr Phe Asp Tyr Trp Gly
115 120 125
Gln Gly Thr Leu Val Thr Val Ser Ser Ala Ser Thr Lys Gly Pro Ser
130 135 140
Val Phe Pro Leu Ala Pro Cys Ser Arg Ser Thr Ser Glu Ser Thr Ala
145 150 155 160
Ala Leu Gly Cys Leu Val Lys Asp Tyr Phe Pro Glu Pro Val Thr Val
165 170 175
Ser Trp Asn Ser Gly Ala Leu Thr Ser Gly Val His Thr Phe Pro Ala
180 185 190
Val Leu Gln Ser Ser Gly Leu Tyr Ser Leu Ser Ser Val Val Thr Val
195 200 205
Pro Ser Ser Ser Leu Gly Thr Lys Thr Tyr Thr Cys Asn Val Asp His
210 215 220
Lys Pro Ser Asn Thr Lys Val Asp Lys Arg Val Glu Ser Lys Tyr Gly
225 230 235 240
Pro Pro Cys Pro Pro Cys Pro Ala Pro Glu Phe Leu Gly Gly Pro Ser
245 250 255
Val Phe Leu Phe Pro Pro Lys Pro Lys Asp Thr Leu Met Ile Ser Arg
260 265 270
Thr Pro Glu Val Thr Cys Val Val Val Asp Val Ser Gln Glu Asp Pro
275 280 285
Glu Val Gln Phe Asn Trp Tyr Val Asp Gly Val Glu Val His Asn Ala
290 295 300
Lys Thr Lys Pro Arg Glu Glu Gln Phe Asn Ser Thr Tyr Arg Val Val
305 310 315 320
Ser Val Leu Thr Val Leu His Gln Asp Trp Leu Asn Gly Lys Glu Tyr
325 330 335
Lys Cys Lys Val Ser Asn Lys Gly Leu Pro Ser Ser Ile Glu Lys Thr
340 345 350
Ile Ser Lys Ala Lys Gly Gln Pro Arg Glu Pro Gln Val Tyr Thr Leu
355 360 365
Pro Pro Ser Gln Glu Glu Met Thr Lys Asn Gln Val Ser Leu Thr Cys
370 375 380
Leu Val Lys Gly Phe Tyr Pro Ser Asp Ile Ala Val Glu Trp Glu Ser
385 390 395 400
Asn Gly Gln Pro Glu Asn Asn Tyr Lys Thr Thr Pro Pro Val Leu Asp
405 410 415
Ser Asp Gly Ser Phe Phe Leu Tyr Ser Arg Leu Thr Val Asp Lys Ser
420 425 430
Arg Trp Gln Glu Gly Asn Val Phe Ser Cys Ser Val Met His Glu Ala
435 440 445
Leu His Asn His Tyr Thr Gln Lys Ser Leu Ser Leu Ser Leu Gly Lys
450 455 460
<210> 58
<211> 233
<212> PRT
<213> Artificial (Artifical)
<220>
<223> light chain-19 aa leader sequence of humanized antibody binding to PD-L1
<400> 58
Met Gly Trp Ser Trp Ile Leu Leu Phe Leu Leu Ser Val Thr Ala Gly
1 5 10 15
Val His Ser Asp Ile Gln Leu Thr Gln Ser Pro Ser Ser Leu Ser Ala
20 25 30
Ser Val Gly Asp Arg Val Thr Ile Thr Cys Lys Thr Ser Gln Asp Val
35 40 45
Asn Thr Ala Val Ala Trp Tyr Gln Gln Lys Pro Gly Gln Ala Pro Arg
50 55 60
Leu Leu Ile Tyr Trp Ala Ser Thr Arg His Thr Gly Val Pro Ser Arg
65 70 75 80
Phe Ser Gly Ser Gly Ser Gly Thr Asp Phe Thr Leu Thr Ile Ser Ser
85 90 95
Leu Gln Pro Glu Asp Phe Ala Thr Tyr Tyr Cys Gln Gln His Tyr Asn
100 105 110
Thr Pro Leu Thr Phe Gly Gly Gly Thr Lys Leu Glu Ile Lys Arg Thr
115 120 125
Val Ala Ala Pro Ser Val Phe Ile Phe Pro Pro Ser Asp Glu Gln Leu
130 135 140
Lys Ser Gly Thr Ala Ser Val Val Cys Leu Leu Asn Asn Phe Tyr Pro
145 150 155 160
Arg Glu Ala Lys Val Gln Trp Lys Val Asp Asn Ala Leu Gln Ser Gly
165 170 175
Asn Ser Gln Glu Ser Val Thr Glu Gln Asp Ser Lys Asp Ser Thr Tyr
180 185 190
Ser Leu Ser Ser Thr Leu Thr Leu Ser Lys Ala Asp Tyr Glu Lys His
195 200 205
Lys Val Tyr Ala Cys Glu Val Thr His Gln Gly Leu Ser Ser Pro Val
210 215 220
Thr Lys Ser Phe Asn Arg Gly Glu Cys
225 230
<210> 59
<211> 464
<212> PRT
<213> Artificial (Artifical)
<220>
<223> heavy chain-19 aa leader sequence of humanized antibody binding to PD-L1
<400> 59
Met Gly Trp Ser Trp Ile Leu Leu Phe Leu Leu Ser Val Thr Ala Gly
1 5 10 15
Val His Ser Gln Met Gln Leu Val Gln Ser Gly Ala Glu Val Lys Lys
20 25 30
Thr Gly Ser Ser Val Lys Val Ser Cys Lys Ala Ser Gly Tyr Thr Phe
35 40 45
Thr Ser Tyr Ser Ile Asn Trp Val Arg Gln Ala Pro Gly Lys Gly Leu
50 55 60
Glu Trp Val Ala Tyr Phe Tyr Val Gly Asn Gly Tyr Thr Asp Tyr Asn
65 70 75 80
Glu Lys Phe Lys Gly Arg Val Thr Met Thr Arg Asp Thr Ser Thr Ser
85 90 95
Thr Val Tyr Met Glu Leu Ser Ser Leu Arg Ser Glu Asp Thr Ala Val
100 105 110
Tyr Tyr Cys Ala Arg Gly Gly Leu Pro Tyr Tyr Phe Asp Tyr Trp Gly
115 120 125
Gln Gly Thr Leu Val Thr Val Ser Ser Ala Ser Thr Lys Gly Pro Ser
130 135 140
Val Phe Pro Leu Ala Pro Cys Ser Arg Ser Thr Ser Glu Ser Thr Ala
145 150 155 160
Ala Leu Gly Cys Leu Val Lys Asp Tyr Phe Pro Glu Pro Val Thr Val
165 170 175
Ser Trp Asn Ser Gly Ala Leu Thr Ser Gly Val His Thr Phe Pro Ala
180 185 190
Val Leu Gln Ser Ser Gly Leu Tyr Ser Leu Ser Ser Val Val Thr Val
195 200 205
Pro Ser Ser Ser Leu Gly Thr Lys Thr Tyr Thr Cys Asn Val Asp His
210 215 220
Lys Pro Ser Asn Thr Lys Val Asp Lys Arg Val Glu Ser Lys Tyr Gly
225 230 235 240
Pro Pro Cys Pro Pro Cys Pro Ala Pro Glu Phe Leu Gly Gly Pro Ser
245 250 255
Val Phe Leu Phe Pro Pro Lys Pro Lys Asp Thr Leu Met Ile Ser Arg
260 265 270
Thr Pro Glu Val Thr Cys Val Val Val Asp Val Ser Gln Glu Asp Pro
275 280 285
Glu Val Gln Phe Asn Trp Tyr Val Asp Gly Val Glu Val His Asn Ala
290 295 300
Lys Thr Lys Pro Arg Glu Glu Gln Phe Asn Ser Thr Tyr Arg Val Val
305 310 315 320
Ser Val Leu Thr Val Leu His Gln Asp Trp Leu Asn Gly Lys Glu Tyr
325 330 335
Lys Cys Lys Val Ser Asn Lys Gly Leu Pro Ser Ser Ile Glu Lys Thr
340 345 350
Ile Ser Lys Ala Lys Gly Gln Pro Arg Glu Pro Gln Val Tyr Thr Leu
355 360 365
Pro Pro Ser Gln Glu Glu Met Thr Lys Asn Gln Val Ser Leu Thr Cys
370 375 380
Leu Val Lys Gly Phe Tyr Pro Ser Asp Ile Ala Val Glu Trp Glu Ser
385 390 395 400
Asn Gly Gln Pro Glu Asn Asn Tyr Lys Thr Thr Pro Pro Val Leu Asp
405 410 415
Ser Asp Gly Ser Phe Phe Leu Tyr Ser Arg Leu Thr Val Asp Lys Ser
420 425 430
Arg Trp Gln Glu Gly Asn Val Phe Ser Cys Ser Val Met His Glu Ala
435 440 445
Leu His Asn His Tyr Thr Gln Lys Ser Leu Ser Leu Ser Leu Gly Lys
450 455 460
<210> 60
<211> 233
<212> PRT
<213> Artificial (Artifical)
<220>
<223> light chain-19 aa leader sequence of humanized antibody binding to PD-L1
<400> 60
Met Gly Trp Ser Trp Ile Leu Leu Phe Leu Leu Ser Val Thr Ala Gly
1 5 10 15
Val His Ser Asp Ile Gln Met Thr Gln Ser Pro Ser Ser Val Ser Ala
20 25 30
Ser Val Gly Asp Arg Val Thr Ile Thr Cys Lys Thr Ser Gln Asp Val
35 40 45
Asn Thr Ala Val Ala Trp Tyr Gln Gln Lys Pro Gly Gln Ala Pro Arg
50 55 60
Leu Leu Ile Tyr Trp Ala Ser Thr Arg His Thr Gly Val Pro Asp Arg
65 70 75 80
Phe Ser Gly Ser Gly Ser Gly Thr Asp Phe Thr Leu Thr Ile Ser Ser
85 90 95
Leu Gln Ala Glu Asp Val Ala Val Tyr Tyr Cys Gln Gln His Tyr Asn
100 105 110
Thr Pro Leu Thr Phe Gly Gly Gly Thr Lys Leu Glu Ile Lys Arg Thr
115 120 125
Val Ala Ala Pro Ser Val Phe Ile Phe Pro Pro Ser Asp Glu Gln Leu
130 135 140
Lys Ser Gly Thr Ala Ser Val Val Cys Leu Leu Asn Asn Phe Tyr Pro
145 150 155 160
Arg Glu Ala Lys Val Gln Trp Lys Val Asp Asn Ala Leu Gln Ser Gly
165 170 175
Asn Ser Gln Glu Ser Val Thr Glu Gln Asp Ser Lys Asp Ser Thr Tyr
180 185 190
Ser Leu Ser Ser Thr Leu Thr Leu Ser Lys Ala Asp Tyr Glu Lys His
195 200 205
Lys Val Tyr Ala Cys Glu Val Thr His Gln Gly Leu Ser Ser Pro Val
210 215 220
Thr Lys Ser Phe Asn Arg Gly Glu Cys
225 230
<210> 61
<211> 464
<212> PRT
<213> Artificial (Artifical)
<220>
<223> heavy chain-19 aa leader sequence of humanized antibody binding to PD-L1
<400> 61
Met Gly Trp Ser Trp Ile Leu Leu Phe Leu Leu Ser Val Thr Ala Gly
1 5 10 15
Val His Ser Gln Val Gln Leu Val Gln Ser Gly Ala Glu Val Lys Lys
20 25 30
Pro Gly Ala Ser Val Lys Val Ser Cys Lys Ala Ser Gly Tyr Thr Phe
35 40 45
Thr Ser Tyr Ser Ile Asn Trp Val Arg Gln Ala Pro Gly Lys Gly Leu
50 55 60
Glu Trp Val Ala Tyr Phe Tyr Val Gly Asn Gly Tyr Thr Asp Tyr Asn
65 70 75 80
Glu Lys Phe Lys Gly Arg Val Thr Ile Thr Thr Asp Glu Ser Thr Ser
85 90 95
Thr Ala Tyr Met Glu Leu Ser Ser Leu Arg Ser Glu Asp Thr Ala Val
100 105 110
Tyr Tyr Cys Ala Arg Gly Gly Leu Pro Tyr Tyr Phe Asp Tyr Trp Gly
115 120 125
Gln Gly Thr Leu Val Thr Val Ser Ser Ala Ser Thr Lys Gly Pro Ser
130 135 140
Val Phe Pro Leu Ala Pro Cys Ser Arg Ser Thr Ser Glu Ser Thr Ala
145 150 155 160
Ala Leu Gly Cys Leu Val Lys Asp Tyr Phe Pro Glu Pro Val Thr Val
165 170 175
Ser Trp Asn Ser Gly Ala Leu Thr Ser Gly Val His Thr Phe Pro Ala
180 185 190
Val Leu Gln Ser Ser Gly Leu Tyr Ser Leu Ser Ser Val Val Thr Val
195 200 205
Pro Ser Ser Ser Leu Gly Thr Lys Thr Tyr Thr Cys Asn Val Asp His
210 215 220
Lys Pro Ser Asn Thr Lys Val Asp Lys Arg Val Glu Ser Lys Tyr Gly
225 230 235 240
Pro Pro Cys Pro Pro Cys Pro Ala Pro Glu Phe Leu Gly Gly Pro Ser
245 250 255
Val Phe Leu Phe Pro Pro Lys Pro Lys Asp Thr Leu Met Ile Ser Arg
260 265 270
Thr Pro Glu Val Thr Cys Val Val Val Asp Val Ser Gln Glu Asp Pro
275 280 285
Glu Val Gln Phe Asn Trp Tyr Val Asp Gly Val Glu Val His Asn Ala
290 295 300
Lys Thr Lys Pro Arg Glu Glu Gln Phe Asn Ser Thr Tyr Arg Val Val
305 310 315 320
Ser Val Leu Thr Val Leu His Gln Asp Trp Leu Asn Gly Lys Glu Tyr
325 330 335
Lys Cys Lys Val Ser Asn Lys Gly Leu Pro Ser Ser Ile Glu Lys Thr
340 345 350
Ile Ser Lys Ala Lys Gly Gln Pro Arg Glu Pro Gln Val Tyr Thr Leu
355 360 365
Pro Pro Ser Gln Glu Glu Met Thr Lys Asn Gln Val Ser Leu Thr Cys
370 375 380
Leu Val Lys Gly Phe Tyr Pro Ser Asp Ile Ala Val Glu Trp Glu Ser
385 390 395 400
Asn Gly Gln Pro Glu Asn Asn Tyr Lys Thr Thr Pro Pro Val Leu Asp
405 410 415
Ser Asp Gly Ser Phe Phe Leu Tyr Ser Arg Leu Thr Val Asp Lys Ser
420 425 430
Arg Trp Gln Glu Gly Asn Val Phe Ser Cys Ser Val Met His Glu Ala
435 440 445
Leu His Asn His Tyr Thr Gln Lys Ser Leu Ser Leu Ser Leu Gly Lys
450 455 460
<210> 62
<211> 233
<212> PRT
<213> Artificial (Artifical)
<220>
<223> light chain-19 aa leader sequence of humanized antibody binding to PD-L1
<400> 62
Met Gly Trp Ser Trp Ile Leu Leu Phe Leu Leu Ser Val Thr Ala Gly
1 5 10 15
Val His Ser Asp Ile Gln Met Thr Gln Ser Pro Ser Ser Leu Ser Ala
20 25 30
Ser Val Gly Asp Arg Val Thr Ile Thr Cys Lys Thr Ser Gln Asp Val
35 40 45
Asn Thr Ala Val Ala Trp Tyr Gln Gln Lys Pro Gly Gln Pro Pro Lys
50 55 60
Leu Leu Ile Tyr Trp Ala Ser Thr Arg His Thr Gly Ile Pro Ala Arg
65 70 75 80
Phe Ser Gly Ser Gly Ser Gly Thr Asp Phe Thr Leu Thr Ile Ser Ser
85 90 95
Leu Gln Pro Glu Asp Phe Ala Val Tyr Tyr Cys Gln Gln His Tyr Asn
100 105 110
Thr Pro Leu Thr Phe Gly Gly Gly Thr Lys Leu Glu Ile Lys Arg Thr
115 120 125
Val Ala Ala Pro Ser Val Phe Ile Phe Pro Pro Ser Asp Glu Gln Leu
130 135 140
Lys Ser Gly Thr Ala Ser Val Val Cys Leu Leu Asn Asn Phe Tyr Pro
145 150 155 160
Arg Glu Ala Lys Val Gln Trp Lys Val Asp Asn Ala Leu Gln Ser Gly
165 170 175
Asn Ser Gln Glu Ser Val Thr Glu Gln Asp Ser Lys Asp Ser Thr Tyr
180 185 190
Ser Leu Ser Ser Thr Leu Thr Leu Ser Lys Ala Asp Tyr Glu Lys His
195 200 205
Lys Val Tyr Ala Cys Glu Val Thr His Gln Gly Leu Ser Ser Pro Val
210 215 220
Thr Lys Ser Phe Asn Arg Gly Glu Cys
225 230
<210> 63
<211> 464
<212> PRT
<213> Artificial (Artifical)
<220>
<223> heavy chain-19 aa leader sequence of humanized antibody binding to PD-L1
<400> 63
Met Gly Trp Ser Trp Ile Leu Leu Phe Leu Leu Ser Val Thr Ala Gly
1 5 10 15
Val His Ser Gln Val Gln Leu Val Gln Ser Gly Ser Glu Leu Lys Lys
20 25 30
Pro Gly Ala Ser Val Lys Val Ser Cys Lys Ala Ser Gly Tyr Thr Phe
35 40 45
Thr Ser Tyr Ser Ile Asn Trp Val Arg Gln Ala Pro Gly Lys Gly Leu
50 55 60
Glu Trp Val Ala Tyr Phe Tyr Val Gly Asn Gly Tyr Thr Asp Tyr Asn
65 70 75 80
Glu Lys Phe Lys Gly Arg Val Thr Ile Thr Ala Asp Glu Ser Thr Ser
85 90 95
Thr Ala Tyr Met Glu Leu Ser Ser Leu Arg Ser Glu Asp Thr Ala Val
100 105 110
Tyr Tyr Cys Ala Arg Gly Gly Leu Pro Tyr Tyr Phe Asp Tyr Trp Gly
115 120 125
Gln Gly Thr Leu Val Thr Val Ser Ser Ala Ser Thr Lys Gly Pro Ser
130 135 140
Val Phe Pro Leu Ala Pro Cys Ser Arg Ser Thr Ser Glu Ser Thr Ala
145 150 155 160
Ala Leu Gly Cys Leu Val Lys Asp Tyr Phe Pro Glu Pro Val Thr Val
165 170 175
Ser Trp Asn Ser Gly Ala Leu Thr Ser Gly Val His Thr Phe Pro Ala
180 185 190
Val Leu Gln Ser Ser Gly Leu Tyr Ser Leu Ser Ser Val Val Thr Val
195 200 205
Pro Ser Ser Ser Leu Gly Thr Lys Thr Tyr Thr Cys Asn Val Asp His
210 215 220
Lys Pro Ser Asn Thr Lys Val Asp Lys Arg Val Glu Ser Lys Tyr Gly
225 230 235 240
Pro Pro Cys Pro Pro Cys Pro Ala Pro Glu Phe Leu Gly Gly Pro Ser
245 250 255
Val Phe Leu Phe Pro Pro Lys Pro Lys Asp Thr Leu Met Ile Ser Arg
260 265 270
Thr Pro Glu Val Thr Cys Val Val Val Asp Val Ser Gln Glu Asp Pro
275 280 285
Glu Val Gln Phe Asn Trp Tyr Val Asp Gly Val Glu Val His Asn Ala
290 295 300
Lys Thr Lys Pro Arg Glu Glu Gln Phe Asn Ser Thr Tyr Arg Val Val
305 310 315 320
Ser Val Leu Thr Val Leu His Gln Asp Trp Leu Asn Gly Lys Glu Tyr
325 330 335
Lys Cys Lys Val Ser Asn Lys Gly Leu Pro Ser Ser Ile Glu Lys Thr
340 345 350
Ile Ser Lys Ala Lys Gly Gln Pro Arg Glu Pro Gln Val Tyr Thr Leu
355 360 365
Pro Pro Ser Gln Glu Glu Met Thr Lys Asn Gln Val Ser Leu Thr Cys
370 375 380
Leu Val Lys Gly Phe Tyr Pro Ser Asp Ile Ala Val Glu Trp Glu Ser
385 390 395 400
Asn Gly Gln Pro Glu Asn Asn Tyr Lys Thr Thr Pro Pro Val Leu Asp
405 410 415
Ser Asp Gly Ser Phe Phe Leu Tyr Ser Arg Leu Thr Val Asp Lys Ser
420 425 430
Arg Trp Gln Glu Gly Asn Val Phe Ser Cys Ser Val Met His Glu Ala
435 440 445
Leu His Asn His Tyr Thr Gln Lys Ser Leu Ser Leu Ser Leu Gly Lys
450 455 460
<210> 64
<211> 233
<212> PRT
<213> Artificial (Artifical)
<220>
<223> light chain-19 aa leader sequence of humanized antibody binding to PD-L1
<400> 64
Met Gly Trp Ser Trp Ile Leu Leu Phe Leu Leu Ser Val Thr Ala Gly
1 5 10 15
Val His Ser Asp Ile Gln Met Thr Gln Ser Pro Ser Thr Leu Ser Ala
20 25 30
Ser Val Gly Asp Arg Val Thr Ile Thr Cys Lys Thr Ser Gln Asp Val
35 40 45
Asn Thr Ala Val Ala Trp Tyr Gln Gln Lys Pro Gly Lys Ala Pro Lys
50 55 60
Leu Leu Leu Tyr Trp Ala Ser Thr Arg His Thr Gly Ile Pro Ala Arg
65 70 75 80
Phe Ser Gly Ser Gly Ser Gly Thr Glu Phe Thr Leu Thr Ile Ser Ser
85 90 95
Leu Gln Ser Glu Asp Phe Ala Val Tyr Tyr Cys Gln Gln His Tyr Asn
100 105 110
Thr Pro Leu Thr Phe Gly Gly Gly Thr Lys Leu Glu Ile Lys Arg Thr
115 120 125
Val Ala Ala Pro Ser Val Phe Ile Phe Pro Pro Ser Asp Glu Gln Leu
130 135 140
Lys Ser Gly Thr Ala Ser Val Val Cys Leu Leu Asn Asn Phe Tyr Pro
145 150 155 160
Arg Glu Ala Lys Val Gln Trp Lys Val Asp Asn Ala Leu Gln Ser Gly
165 170 175
Asn Ser Gln Glu Ser Val Thr Glu Gln Asp Ser Lys Asp Ser Thr Tyr
180 185 190
Ser Leu Ser Ser Thr Leu Thr Leu Ser Lys Ala Asp Tyr Glu Lys His
195 200 205
Lys Val Tyr Ala Cys Glu Val Thr His Gln Gly Leu Ser Ser Pro Val
210 215 220
Thr Lys Ser Phe Asn Arg Gly Glu Cys
225 230
<210> 65
<211> 1392
<212> DNA
<213> Artificial (Artifical)
<220>
<223> heavy chain-19 aa leader sequence of humanized antibody binding to PD-L1
<400> 65
atgggctgga gctggatcct gctgttcctc ctgagcgtga cagcaggagt gcacagcgag 60
gtgcagctgg tgcagagcgg ggcagaggtc aaaaagcctg gcgaaagcct gaaaatctcc 120
tgtaagggaa gcggatactc attcacctct tacagcatca actgggtgcg gcagatgcca 180
ggcaagggcc tggagtggat gggctacttc tatgtgggca acggctacac agactataat 240
gagaagttta agggccgggt gaccatgaca agaaatacct ccatctctac agcctatatg 300
gagctgagct ccctgaggag cgaggatacc gccgtgtact attgcgcccg cggggggctg 360
ccatactatt ttgactattg gggacagggg actctggtga ccgtctcatc cgcctctaca 420
aagggcccct ccgtgtttcc actggctccc tgcagcaggt ctacatccga gagcaccgct 480
gctctgggat gtctggtgaa ggattacttc cctgagccag tgaccgtgag ctggaactcc 540
ggagctctga catccggagt gcacaccttt cctgctgtgc tgcagagctc tggcctgtac 600
agcctgtcca gcgtggtgac agtgccatct tccagcctgg gcaccaagac atatacctgc 660
aacgtggacc ataagcccag caataccaag gtggataaga gagtggagtc taagtacgga 720
ccaccttgcc caccatgtcc agctcctgag tttctgggag gaccatccgt gttcctgttt 780
cctccaaagc ctaaggacac cctgatgatc tctcgcacac ccgaggtgac ctgtgtggtg 840
gtggacgtgt cccaggagga tcctgaggtg cagttcaact ggtacgtgga tggcgtggag 900
gtgcacaatg ctaagaccaa gcctagggag gagcagttta acagcacata ccgggtggtg 960
tctgtgctga ccgtgctgca tcaggactgg ctgaacggca aggagtataa gtgcaaggtg 1020
agcaataagg gcctgccatc ttccatcgag aagacaatct ctaaggctaa gggacagcct 1080
agggagccac aggtgtacac cctgccccct tcccaggagg agatgacaaa gaaccaggtg 1140
agcctgacct gtctggtgaa gggcttctat ccttctgaca tcgctgtgga gtgggagtcc 1200
aatggccagc cagagaacaa ttacaagacc acaccacccg tgctggactc cgatggcagc 1260
ttctttctgt attccaggct gaccgtggat aagagccggt ggcaggaggg caatgtgttt 1320
tcttgttccg tgatgcacga agcactgcac aaccactaca ctcagaagtc cctgtcactg 1380
tccctgggca ag 1392
<210> 66
<211> 699
<212> DNA
<213> Artificial (Artifical)
<220>
<223> light chain-19 aa leader sequence of humanized antibody binding to PD-L1
<400> 66
atgggctgga gctggatcct gctgttcctc ctgagcgtga cagcaggagt gcacagcgac 60
attcagatga cacagagccc ttcaacactg agcgcctccg tgggggatcg ggtgactatt 120
acttgtaaga ccagccagga cgtgaacacc gcagtggcat ggtaccagca gaagccaggc 180
aaggccccta agctgctgat ctattgggcc tctacccggc acacaggcgt gccaagcaga 240
ttctctggca gcggctccgg caccgagttt accctgacaa tcagctccct gcagcccgag 300
gacttcgcca catactattg ccagcagcat tacaacacac ccctgacatt cggcggcggg 360
acaaaactgg aaatcaagag gaccgtggcc gctcccagtg tcttcatttt tccccctagc 420
gacgaacagc tgaaatccgg gactgcttct gtggtctgtc tgctgaacaa tttctaccct 480
cgcgaagcca aagtgcagtg gaaggtcgat aacgctctcc agagtggcaa ttcacaggag 540
agcgtgacag aacaggactc caaagattct acttatagtc tgtctagtac actgactctg 600
tccaaggcag actacgagaa gcacaaagtg tatgcctgtg aagtcaccca tcagggcctg 660
tcaagccccg tgacaaagtc ttttaacaga ggggagtgt 699
<210> 67
<211> 1392
<212> DNA
<213> Artificial (Artifical)
<220>
<223> heavy chain-19 aa leader sequence of humanized antibody binding to PD-L1
<400> 67
atgggctgga gctggatcct gctgttcctc ctgagcgtga cagcaggagt gcacagcgag 60
gtgcagctgg tgcagagcgg ggcagaagtc aagaagcctg gggaaagcct gaaaatctcc 120
tgtaagggaa gcggatactc attcacctct tacagcatca actgggtgcg gcaggcacca 180
ggcaagggcc tggagtggat gggctacttc tatgtgggca acggctacac agactataat 240
gagaagttta agggccgggt gaccatgaca agaaatacct ccatctctac agcctatatg 300
gagctgagct ccctgaggag cgaggatacc gccgtgtact attgcgcccg cgggggactg 360
ccttactatt ttgactactg gggacaggga acactggtga ccgtctcctc tgcctctaca 420
aagggcccct ccgtgtttcc actggctccc tgcagcaggt ctacatccga gagcaccgct 480
gctctgggat gtctggtgaa ggattacttc cctgagccag tgaccgtgag ctggaactcc 540
ggagctctga catccggagt gcacaccttt cctgctgtgc tgcagagctc tggcctgtac 600
agcctgtcca gcgtggtgac agtgccatct tccagcctgg gcaccaagac atatacctgc 660
aacgtggacc ataagcccag caataccaag gtggataaga gagtggagtc taagtacgga 720
ccaccttgcc caccatgtcc agctcctgag tttctgggag gaccatccgt gttcctgttt 780
cctccaaagc ctaaggacac cctgatgatc tctcgcacac ccgaggtgac ctgtgtggtg 840
gtggacgtgt cccaggagga tcctgaggtg cagttcaact ggtacgtgga tggcgtggag 900
gtgcacaatg ctaagaccaa gcctagggag gagcagttta acagcacata ccgggtggtg 960
tctgtgctga ccgtgctgca tcaggactgg ctgaacggca aggagtataa gtgcaaggtg 1020
agcaataagg gcctgccatc ttccatcgag aagacaatct ctaaggctaa gggacagcct 1080
agggagccac aggtgtacac cctgccccct tcccaggagg agatgacaaa gaaccaggtg 1140
agcctgacct gtctggtgaa gggcttctat ccttctgaca tcgctgtgga gtgggagtcc 1200
aatggccagc cagagaacaa ttacaagacc acaccacccg tgctggactc cgatggcagc 1260
ttctttctgt attccaggct gaccgtggat aagagccggt ggcaggaggg caatgtgttt 1320
tcttgttccg tgatgcacga agcactgcac aaccactaca ctcagaagtc cctgtcactg 1380
tccctgggca ag 1392
<210> 68
<211> 699
<212> DNA
<213> Artificial (Artifical)
<220>
<223> light chain-19 aa leader sequence of humanized antibody binding to PD-L1
<400> 68
atgggctgga gctggatcct gctgttcctc ctgagcgtga cagcaggagt gcacagcgac 60
atccagctga cccagagccc aagtagcctg agcgcaagcg tcggggacag agtgactatt 120
acctgcaaga caagccagga cgtgaacacc gcagtggcat ggtaccagca gaagccagga 180
caggcaccta ggctgctgat ctattgggcc tctacccggc acacaggcgt gccaagcaga 240
ttctctggca gcggctccgg caccgacttt accctgacaa tcagctccct gcagcccgag 300
gatttcgcca catactattg ccagcagcat tacaacactc ctctgacatt cggggggggg 360
acaaaactgg aaatcaaaag gaccgtggcc gctcccagtg tcttcatttt tccccctagc 420
gacgaacagc tgaaatccgg gactgcttct gtggtctgtc tgctgaacaa tttctaccct 480
cgcgaagcca aagtgcagtg gaaggtcgat aacgctctcc agagtggcaa ttcacaggag 540
agcgtgacag aacaggactc caaagattct acttatagtc tgtctagtac actgactctg 600
tccaaggcag actacgagaa gcacaaagtg tatgcctgtg aagtcaccca tcagggcctg 660
tcaagccccg tgacaaagtc ttttaacaga ggggagtgt 699
<210> 69
<211> 1392
<212> DNA
<213> Artificial (Artifical)
<220>
<223> heavy chain-19 aa leader sequence of humanized antibody binding to PD-L1
<400> 69
atgggctgga gctggatcct gctgttcctc ctgagcgtga cagcaggagt gcacagccag 60
atgcagctgg tccagtcagg cgcagaagtc aagaaaaccg ggtcaagcgt caaagtgtca 120
tgtaaagcaa gcggatatac cttcacatct tacagcatca actgggtgcg gcaggcacca 180
ggcaagggcc tggagtgggt ggcctacttc tatgtgggca acggctacac cgactataat 240
gagaagttta agggccgggt gaccatgaca agagatacct ccacatctac cgtgtatatg 300
gagctgagct ccctgaggag cgaggacaca gccgtgtact attgcgcccg cggggggctg 360
ccatactact ttgactattg gggacaggga acactggtga ccgtctcatc agcctctaca 420
aagggcccct ccgtgtttcc actggctccc tgcagcaggt ctacatccga gagcaccgct 480
gctctgggat gtctggtgaa ggattacttc cctgagccag tgaccgtgag ctggaactcc 540
ggagctctga catccggagt gcacaccttt cctgctgtgc tgcagagctc tggcctgtac 600
agcctgtcca gcgtggtgac agtgccatct tccagcctgg gcaccaagac atatacctgc 660
aacgtggacc ataagcccag caataccaag gtggataaga gagtggagtc taagtacgga 720
ccaccttgcc caccatgtcc agctcctgag tttctgggag gaccatccgt gttcctgttt 780
cctccaaagc ctaaggacac cctgatgatc tctcgcacac ccgaggtgac ctgtgtggtg 840
gtggacgtgt cccaggagga tcctgaggtg cagttcaact ggtacgtgga tggcgtggag 900
gtgcacaatg ctaagaccaa gcctagggag gagcagttta acagcacata ccgggtggtg 960
tctgtgctga ccgtgctgca tcaggactgg ctgaacggca aggagtataa gtgcaaggtg 1020
agcaataagg gcctgccatc ttccatcgag aagacaatct ctaaggctaa gggacagcct 1080
agggagccac aggtgtacac cctgccccct tcccaggagg agatgacaaa gaaccaggtg 1140
agcctgacct gtctggtgaa gggcttctat ccttctgaca tcgctgtgga gtgggagtcc 1200
aatggccagc cagagaacaa ttacaagacc acaccacccg tgctggactc cgatggcagc 1260
ttctttctgt attccaggct gaccgtggat aagagccggt ggcaggaggg caatgtgttt 1320
tcttgttccg tgatgcacga agcactgcac aaccactaca ctcagaagtc cctgtcactg 1380
tccctgggca ag 1392
<210> 70
<211> 699
<212> DNA
<213> Artificial (Artifical)
<220>
<223> light chain-19 aa leader sequence of humanized antibody binding to PD-L1
<400> 70
atgggctgga gctggatcct gctgttcctc ctgagcgtga cagcaggagt gcacagcgac 60
attcagatga cccagtctcc aagctcagtc tcagccagtg tgggcgaccg agtgacaatt 120
acatgcaaaa caagccagga cgtgaacacc gcagtggcat ggtaccagca gaagccagga 180
caggcaccta ggctgctgat ctattgggca tctaccaggc acacaggagt gccagacaga 240
ttctctggca gcggctccgg cacagatttt accctgacaa tcagctccct gcaggcagag 300
gacgtggcag tgtactattg ccagcagcat tacaatactc ctctgacatt cgggggcgga 360
accaaactgg aaatcaaaag gaccgtggcc gctcccagtg tcttcatttt tccccctagc 420
gacgaacagc tgaaatccgg gactgcttct gtggtctgtc tgctgaacaa tttctaccct 480
cgcgaagcca aagtgcagtg gaaggtcgat aacgctctcc agagtggcaa ttcacaggag 540
agcgtgacag aacaggactc caaagattct acttatagtc tgtctagtac actgactctg 600
tccaaggcag actacgagaa gcacaaagtg tatgcctgtg aagtcaccca tcagggcctg 660
tcaagccccg tgacaaagtc ttttaacaga ggggagtgt 699
<210> 71
<211> 1392
<212> DNA
<213> Artificial (Artifical)
<220>
<223> heavy chain-19 aa leader sequence of humanized antibody binding to PD-L1
<400> 71
atgggctgga gctggatcct gctgttcctc ctgagcgtga cagcaggagt gcacagccag 60
gtccagctgg tccagtcagg ggcagaagtg aaaaaacccg gagcctcagt caaagtgtca 120
tgcaaagcaa gcggatacac atttacctct tacagcatca actgggtgag gcaggcacca 180
ggcaagggcc tggagtgggt ggcctacttc tatgtgggca acggctacac cgactataat 240
gagaagttta agggccgggt gacaatcacc acagatgagt ccacctctac agcctatatg 300
gagctgagct ccctgcggag cgaggacaca gccgtgtact attgcgccag aggcgggctg 360
ccttattatt ttgactattg gggacagggg acactggtga ccgtctcttc agcctctaca 420
aagggcccct ccgtgtttcc actggctccc tgcagcaggt ctacatccga gagcaccgct 480
gctctgggat gtctggtgaa ggattacttc cctgagccag tgaccgtgag ctggaactcc 540
ggagctctga catccggagt gcacaccttt cctgctgtgc tgcagagctc tggcctgtac 600
agcctgtcca gcgtggtgac agtgccatct tccagcctgg gcaccaagac atatacctgc 660
aacgtggacc ataagcccag caataccaag gtggataaga gagtggagtc taagtacgga 720
ccaccttgcc caccatgtcc agctcctgag tttctgggag gaccatccgt gttcctgttt 780
cctccaaagc ctaaggacac cctgatgatc tctcgcacac ccgaggtgac ctgtgtggtg 840
gtggacgtgt cccaggagga tcctgaggtg cagttcaact ggtacgtgga tggcgtggag 900
gtgcacaatg ctaagaccaa gcctagggag gagcagttta acagcacata ccgggtggtg 960
tctgtgctga ccgtgctgca tcaggactgg ctgaacggca aggagtataa gtgcaaggtg 1020
agcaataagg gcctgccatc ttccatcgag aagacaatct ctaaggctaa gggacagcct 1080
agggagccac aggtgtacac cctgccccct tcccaggagg agatgacaaa gaaccaggtg 1140
agcctgacct gtctggtgaa gggcttctat ccttctgaca tcgctgtgga gtgggagtcc 1200
aatggccagc cagagaacaa ttacaagacc acaccacccg tgctggactc cgatggcagc 1260
ttctttctgt attccaggct gaccgtggat aagagccggt ggcaggaggg caatgtgttt 1320
tcttgttccg tgatgcacga agcactgcac aaccactaca ctcagaagtc cctgtcactg 1380
tccctgggca ag 1392
<210> 72
<211> 699
<212> DNA
<213> Artificial (Artifical)
<220>
<223> light chain-19 aa leader sequence of humanized antibody binding to PD-L1
<400> 72
atgggctgga gctggatcct gctgttcctc ctgagcgtga cagcaggagt gcacagcgac 60
attcagatga cacagagccc tagttcactg agcgccagcg tgggagatag agtcacaatc 120
acatgcaaaa caagccagga cgtgaacacc gccgtggcct ggtaccagca gaagccaggc 180
cagcccccta agctgctgat ctattgggca tctaccaggc acacaggcat ccccgcaaga 240
ttctctggaa gcggatccgg cacagacttt accctgacaa tcagctccct gcagcccgag 300
gatttcgccg tgtactattg ccagcagcac tacaatacac cactgacctt cggcggggga 360
actaaactgg aaatcaaaag gaccgtggcc gctcccagtg tcttcatttt tccccctagc 420
gacgaacagc tgaaatccgg gactgcttct gtggtctgtc tgctgaacaa tttctaccct 480
cgcgaagcca aagtgcagtg gaaggtcgat aacgctctcc agagtggcaa ttcacaggag 540
agcgtgacag aacaggactc caaagattct acttatagtc tgtctagtac actgactctg 600
tccaaggcag actacgagaa gcacaaagtg tatgcctgtg aagtcaccca tcagggcctg 660
tcaagccccg tgacaaagtc ttttaacaga ggggagtgt 699
<210> 73
<211> 1392
<212> DNA
<213> Artificial (Artifical)
<220>
<223> heavy chain-19 aa leader sequence of humanized antibody binding to PD-L1
<400> 73
atgggctgga gctggatcct gctgttcctc ctgagcgtga cagcaggagt gcacagccag 60
gtccagctgg tccagagcgg aagtgaactg aagaaacccg gagcaagcgt caaagtctca 120
tgcaaagcaa gcggctacac atttacctct tacagcatca actgggtgag gcaggcacca 180
ggcaagggcc tggagtgggt ggcctacttc tatgtgggca acggctacac agactataat 240
gagaagttta agggccgggt gaccatcaca gccgatgagt ccacctctac agcctatatg 300
gagctgagct ccctgaggag cgaggacacc gccgtgtact attgcgccag aggagggctg 360
ccttattact ttgattactg gggacaggga acactggtga ccgtctcatc cgcctctaca 420
aagggcccct ccgtgtttcc actggctccc tgcagcaggt ctacatccga gagcaccgct 480
gctctgggat gtctggtgaa ggattacttc cctgagccag tgaccgtgag ctggaactcc 540
ggagctctga catccggagt gcacaccttt cctgctgtgc tgcagagctc tggcctgtac 600
agcctgtcca gcgtggtgac agtgccatct tccagcctgg gcaccaagac atatacctgc 660
aacgtggacc ataagcccag caataccaag gtggataaga gagtggagtc taagtacgga 720
ccaccttgcc caccatgtcc agctcctgag tttctgggag gaccatccgt gttcctgttt 780
cctccaaagc ctaaggacac cctgatgatc tctcgcacac ccgaggtgac ctgtgtggtg 840
gtggacgtgt cccaggagga tcctgaggtg cagttcaact ggtacgtgga tggcgtggag 900
gtgcacaatg ctaagaccaa gcctagggag gagcagttta acagcacata ccgggtggtg 960
tctgtgctga ccgtgctgca tcaggactgg ctgaacggca aggagtataa gtgcaaggtg 1020
agcaataagg gcctgccatc ttccatcgag aagacaatct ctaaggctaa gggacagcct 1080
agggagccac aggtgtacac cctgccccct tcccaggagg agatgacaaa gaaccaggtg 1140
agcctgacct gtctggtgaa gggcttctat ccttctgaca tcgctgtgga gtgggagtcc 1200
aatggccagc cagagaacaa ttacaagacc acaccacccg tgctggactc cgatggcagc 1260
ttctttctgt attccaggct gaccgtggat aagagccggt ggcaggaggg caatgtgttt 1320
tcttgttccg tgatgcacga agcactgcac aaccactaca ctcagaagtc cctgtcactg 1380
tccctgggca ag 1392
<210> 74
<211> 699
<212> DNA
<213> Artificial (Artifical)
<220>
<223> light chain-19 aa leader sequence of humanized antibody binding to PD-L1
<400> 74
atgggctgga gctggatcct gctgttcctc ctgagcgtga cagcaggagt gcacagcgat 60
attcagatga cccagagccc aagcaccctg tccgcaagcg tcggggatag agtgaccatt 120
acctgtaaaa caagccagga tgtgaacacc gcagtggcat ggtaccagca gaagccaggc 180
aaggccccta agctgctgct gtattgggca tctaccaggc acacaggcat ccccgcaaga 240
ttctctggaa gcggatccgg cacagagttt accctgacaa tcagctccct gcagagcgag 300
gacttcgccg tgtactattg ccagcagcat tacaatacac cactgacatt cggggggggg 360
actaaactgg aaatcaagag gaccgtggcc gctcccagtg tcttcatttt tccccctagc 420
gacgaacagc tgaaatccgg gactgcttct gtggtctgtc tgctgaacaa tttctaccct 480
cgcgaagcca aagtgcagtg gaaggtcgat aacgctctcc agagtggcaa ttcacaggag 540
agcgtgacag aacaggactc caaagattct acttatagtc tgtctagtac actgactctg 600
tccaaggcag actacgagaa gcacaaagtg tatgcctgtg aagtcaccca tcagggcctg 660
tcaagccccg tgacaaagtc ttttaacaga ggggagtgt 699

Claims (19)

1. An isolated antibody or antigen-binding fragment thereof that specifically binds to human programmed death ligand 1 (PD-L1) and comprises: the light chain CDR1 sequence set forth in SEQ ID NO. 8, the light chain CDR2 sequence set forth in SEQ ID NO. 9, the light chain CDR3 sequence set forth in SEQ ID NO. 11, the heavy chain CDR1 sequence set forth in SEQ ID NO. 2, the heavy chain CDR2 sequence set forth in SEQ ID NO. 4 and the heavy chain CDR3 sequence set forth in SEQ ID NO. 6.
2. An isolated antibody or antigen-binding fragment thereof that specifically binds to human PD-L1 and comprises: (a) The light chain CDR1 sequence set forth in SEQ ID NO. 8, the light chain CDR2 sequence set forth in SEQ ID NO. 9, the light chain CDR3 sequence set forth in SEQ ID NO. 11, the heavy chain CDR1 sequence set forth in SEQ ID NO. 2, the heavy chain CDR2 sequence set forth in SEQ ID NO. 4 and the heavy chain CDR3 sequence set forth in SEQ ID NO. 6; and (b) a set of four variable region framework regions from a human immunoglobulin.
3. The isolated antibody or antigen-binding fragment thereof of claim 2, wherein the human immunoglobulin is IgG.
4. The isolated antibody or antigen-binding fragment thereof of any one of claims 1-3, which is at least 1 x 10 -6 Dissociation constant of M (K D ) Binds to the PD-L1 protein.
5. The isolated antibody or antigen-binding fragment thereof of any one of claims 1-3, which is at least 1 x 10 -7 Dissociation constant of M (K D ) Binds to the PD-L1 protein.
6. The isolated antibody or antigen-binding fragment thereof of any one of claims 1-3, which is at least 1 x 10 -8 Dissociation constant of M (K D ) Binds to the PD-L1 protein.
7. The isolated antibody or antigen-binding fragment thereof of any one of claims 1-3, which is at least 1 x 10 -9 Dissociation constant of M (K D ) Binds to the PD-L1 protein.
8. The isolated antibody or antigen-binding fragment thereof of any one of claims 1-3, which is at least 1.6x10 -10 Dissociation constant of M (K D ) Binds to the PD-L1 protein.
9. An isolated humanized antibody or antigen-binding fragment thereof that specifically binds to human PD-L1 and comprises:
a heavy chain variable region sequence identical to SEQ ID NO. 20 and a light chain variable region sequence identical to SEQ ID NO. 21;
a heavy chain variable region sequence identical to SEQ ID NO. 22 and a light chain variable region sequence identical to SEQ ID NO. 23;
a heavy chain variable region sequence identical to SEQ ID NO. 24 and a light chain variable region sequence identical to SEQ ID NO. 25;
a heavy chain variable region sequence identical to SEQ ID NO. 26 and a light chain variable region sequence identical to SEQ ID NO. 27;
a heavy chain variable region sequence identical to SEQ ID NO. 28 and a light chain variable region sequence identical to SEQ ID NO. 29;
a heavy chain variable region sequence identical to SEQ ID NO. 30 and a light chain variable region sequence identical to SEQ ID NO. 31;
a heavy chain variable region sequence identical to SEQ ID NO. 32 and a light chain variable region sequence identical to SEQ ID NO. 33;
a heavy chain variable region sequence identical to SEQ ID NO. 34 and a light chain variable region sequence identical to SEQ ID NO. 35;
a heavy chain variable region sequence identical to SEQ ID NO. 36 and a light chain variable region sequence identical to SEQ ID NO. 37;
A heavy chain variable region sequence identical to SEQ ID NO. 38 and a light chain variable region sequence identical to SEQ ID NO. 39;
a heavy chain variable region sequence identical to SEQ ID NO. 40 and a light chain variable region sequence identical to SEQ ID NO. 41;
a heavy chain variable region sequence identical to SEQ ID NO. 42 and a light chain variable region sequence identical to SEQ ID NO. 43;
a heavy chain variable region sequence identical to SEQ ID NO. 44 and a light chain variable region sequence identical to SEQ ID NO. 45; or alternatively
A heavy chain variable region sequence identical to SEQ ID NO. 46 and a light chain variable region sequence identical to SEQ ID NO. 47.
10. An isolated humanized antibody or antigen-binding fragment thereof that specifically binds to human PD-L1 and comprises:
the heavy chain sequence set forth in SEQ ID NO. 55 and the light chain sequence set forth in SEQ ID NO. 56;
the heavy chain sequence set forth in SEQ ID NO. 57 and the light chain sequence set forth in SEQ ID NO. 58;
the heavy chain sequence set forth in SEQ ID NO. 59 and the light chain sequence set forth in SEQ ID NO. 60;
the heavy chain sequence set forth in SEQ ID NO. 61 and the light chain sequence set forth in SEQ ID NO. 62; or alternatively
The heavy chain sequence set forth in SEQ ID NO. 63 and the light chain sequence set forth in SEQ ID NO. 64.
11. A pharmaceutical composition comprising the isolated antibody or antigen-binding fragment thereof according to any one of claims 1-10 in admixture with a pharmaceutically acceptable carrier.
12. Use of an isolated antibody or antigen-binding fragment thereof according to any one of claims 1-10 in the manufacture of a pharmaceutical composition for the treatment of a solid tumor, wherein the solid tumor is mediated by PD-L1.
13. Use of an isolated antibody or antigen-binding fragment thereof according to any one of claims 1-10 in the manufacture of a pharmaceutical composition for the treatment of a solid tumor, wherein the solid tumor is mediated by PD-L1, wherein the treatment further comprises one or more additional therapies selected from the group consisting of small molecule kinase inhibitor targeted therapy, surgery, radiation therapy, vaccination regimens and stem cell transplantation, and wherein there is a synergy between the isolated antibody or antigen-binding fragment and the additional therapies when co-administered.
14. Use of an isolated antibody or antigen-binding fragment thereof according to any one of claims 1-10 in the manufacture of a pharmaceutical composition for the treatment of a solid tumor, wherein the solid tumor is mediated by PD-L1, wherein the treatment further comprises immunotherapy, and wherein there is a synergy between the isolated antibody or antigen-binding fragment and the immunotherapy when co-administered.
15. Use of an isolated antibody or antigen-binding fragment thereof according to any one of claims 1-10 in the manufacture of a pharmaceutical composition for the treatment of a solid tumor, wherein the solid tumor is mediated by PD-L1, wherein the treatment further comprises chemotherapy, and wherein there is a synergy between the isolated antibody or antigen-binding fragment and the chemotherapy when co-administered.
16. An isolated immunoconjugate or fusion protein comprising the antibody or antigen-binding fragment thereof according to any one of claims 1-10 coupled to an effector molecule.
17. An isolated nucleic acid comprising a polynucleotide sequence encoding the antibody or antigen-binding fragment thereof according to any one of claims 1-10.
18. A recombinant expression vector comprising the isolated nucleic acid of claim 17.
19. A host cell comprising the vector of claim 18.
CN201780087354.6A 2016-12-23 2017-12-23 Immunotherapy using antibodies that bind to programmed death ligand 1 (PD-L1) Active CN110337448B (en)

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US201662438622P 2016-12-23 2016-12-23
US62/438,622 2016-12-23
PCT/US2017/068369 WO2018119475A1 (en) 2016-12-23 2017-12-23 Immunotherapy using antibodies that bind programmed death ligand-1 (pd-l1)

Publications (2)

Publication Number Publication Date
CN110337448A CN110337448A (en) 2019-10-15
CN110337448B true CN110337448B (en) 2023-08-08

Family

ID=62627185

Family Applications (1)

Application Number Title Priority Date Filing Date
CN201780087354.6A Active CN110337448B (en) 2016-12-23 2017-12-23 Immunotherapy using antibodies that bind to programmed death ligand 1 (PD-L1)

Country Status (6)

Country Link
US (1) US11332531B2 (en)
EP (1) EP3559045A4 (en)
JP (1) JP7128529B2 (en)
CN (1) CN110337448B (en)
RU (1) RU2766582C2 (en)
WO (1) WO2018119475A1 (en)

Families Citing this family (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
GB201403775D0 (en) 2014-03-04 2014-04-16 Kymab Ltd Antibodies, uses & methods
US9567399B1 (en) 2016-06-20 2017-02-14 Kymab Limited Antibodies and immunocytokines
KR20230149857A (en) 2016-07-07 2023-10-27 더 보드 어브 트러스티스 어브 더 리랜드 스탠포드 주니어 유니버시티 Antibody adjuvant conjugates
US11779604B2 (en) 2016-11-03 2023-10-10 Kymab Limited Antibodies, combinations comprising antibodies, biomarkers, uses and methods
AU2018278327B2 (en) 2017-06-01 2023-03-16 Cytomx Therapeutics, Inc. Activatable anti-pdl1 antibodies and methods of use thereof
AU2019241345A1 (en) * 2018-03-29 2020-10-15 Adagene Inc. Anti-PD-L1 antibodies and use thereof
AU2020241686A1 (en) 2019-03-15 2021-11-04 Bolt Biotherapeutics, Inc. Immunoconjugates targeting HER2
IL308918A (en) * 2021-06-09 2024-01-01 Shanghai Epimab Biotherapeutics Co Ltd Antibodies and bispecific binding proteins that bind ox40 and/or pd-l1
CN117126282B (en) * 2023-10-26 2024-01-12 迈威(上海)生物科技股份有限公司 Antibody and application thereof in preparation of medicine for blocking combination of alpha v beta 8 and Latent TGF-beta

Family Cites Families (16)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
AU2001271422B2 (en) * 2000-06-22 2005-12-22 Genentech, Inc. Agonist anti-trk-C monoclonal antibodies
US7514078B2 (en) 2001-06-01 2009-04-07 Cornell Research Foundation, Inc. Methods of treating prostate cancer with anti-prostate specific membrane antigen antibodies
EP1544291A4 (en) 2002-07-19 2006-08-02 Towa Kagaku Co Ltd Novel process for producing antibody enzyme, novel antibody enzyme and utilization thereof
HUE026039T2 (en) * 2005-07-01 2016-05-30 Squibb & Sons Llc Human monoclonal antibodies to programmed death ligand 1 (pd-l1)
KR100882445B1 (en) 2007-03-16 2009-02-09 울산대학교 산학협력단 A method for isolating and expanding autologous antigen-specific CD8+ T cells using an anti-4-1BB antibody
EP2853267B1 (en) 2007-09-21 2016-12-07 The Regents of the University of California Targeted interferon demonstrates potent apoptotic and anti-tumor activities
US20110311550A1 (en) * 2008-10-24 2011-12-22 The Scripps Research Institute Agents for hcv treatment
CN114835812A (en) * 2008-12-09 2022-08-02 霍夫曼-拉罗奇有限公司 anti-PD-L1 antibodies and their use for enhancing T cell function
SG172427A1 (en) * 2008-12-26 2011-07-28 Univ Tokyo Diagnosis and treatment of cancer using anti-lgr7 antibody
JP5725508B2 (en) * 2009-03-25 2015-05-27 国立大学法人東北大学 LH type bispecific antibody
JP2012034668A (en) * 2010-08-12 2012-02-23 Tohoku Univ Fragment of humanized anti-egfr antibody substituted-lysine variable region, and use thereof
WO2012103165A2 (en) * 2011-01-26 2012-08-02 Kolltan Pharmaceuticals, Inc. Anti-kit antibodies and uses thereof
RU2625034C2 (en) * 2011-04-20 2017-07-11 МЕДИММЬЮН, ЭлЭлСи Antibodies and other molecules binding b7-h1 and pd-1
AU2012344260B2 (en) * 2011-11-28 2017-09-07 Merck Patent Gmbh Anti-PD-L1 antibodies and uses thereof
ES2881484T3 (en) * 2014-12-22 2021-11-29 Pd 1 Acquisition Group Llc Anti-PD-1 antibodies
KR20170105622A (en) * 2015-01-26 2017-09-19 마크로제닉스, 인크. A multivalent molecule comprising a DR5-binding domain

Also Published As

Publication number Publication date
EP3559045A1 (en) 2019-10-30
RU2019121086A3 (en) 2021-09-03
RU2766582C2 (en) 2022-03-15
WO2018119475A1 (en) 2018-06-28
EP3559045A4 (en) 2020-08-19
JP7128529B2 (en) 2022-08-31
JP2020514277A (en) 2020-05-21
RU2019121086A (en) 2021-01-25
US20200190197A1 (en) 2020-06-18
CN110337448A (en) 2019-10-15
US11332531B2 (en) 2022-05-17

Similar Documents

Publication Publication Date Title
JP7165995B2 (en) Cancer treatment using antibodies that bind to cytotoxic T lymphocyte antigen-4 (CTLA-4)
CN110337448B (en) Immunotherapy using antibodies that bind to programmed death ligand 1 (PD-L1)
CN110366564B (en) Immunotherapy using antibodies that bind to programmed death 1 (PD-1)
US11242398B2 (en) Anti-OX40 antibodies and methods of activating OX40
KR20140091040A (en) Antibodies specific for trop-2 and their uses
CN113683695A (en) anti-CXCR 4 antibodies and antibody-drug conjugates
US20240101717A1 (en) Anti-her-2/trop-2 constructs and uses thereof
US20230151104A1 (en) Chemokine receptor 4 (cxcr4) antagonist antibodies
CN112040949A (en) Calcitonin Gene Related Peptide (CGRP) antagonist antibodies
US20230235080A1 (en) Trophoblast cell-surface antigen-2 (trop-2) antibodies
WO2024076514A1 (en) C-c chemokine receptor type 8 (ccr8) antagonist antibodies
US20210079087A1 (en) Treatment of autoimmune and inflammatory disorders using antibodies that bind interleukin-17a (il-17a)

Legal Events

Date Code Title Description
PB01 Publication
PB01 Publication
SE01 Entry into force of request for substantive examination
SE01 Entry into force of request for substantive examination
GR01 Patent grant
GR01 Patent grant